Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Anal Chem ; 95(28): 10595-10602, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37403445

RESUMEN

Tandem mass tags (TMT) are one of the most widely used techniques in proteomics quantification due to their ability to accurately and precisely analyze up to 18 samples in a multiplexed manner. Moreover, TMT tags are introduced chemically by covalent coupling of the primary amines of digested proteins, making them universally applicable for any kind of sample. However, in addition to amine groups, the hydroxyl groups of serine, threonine, and tyrosine residues can also be labeled to some extent during TMT labeling, which compromises the analytical sensitivity and results in lower peptide identification rates compared to label-free methods. In this work, we investigated in-depth the chemical nature of TMT overlabeling and revealed that peptides simultaneously containing histidine and hydroxyl-containing residues were prone to overlabeling due to an intramolecular catalysis mediated by the histidyl imidazolyl group. Based on the understanding of the chemical mechanism, we developed a novel TMT labeling method under acidic pH that completely overcomes overlabeling. Compared to the standard labeling method provided by the TMT vendor, our method achieved comparable labeling efficiency on target groups but greatly reduced overlabeled peptides, resulting in the identification of 33.9% more unique peptides and 20.9% more proteins in proteomic analysis.


Asunto(s)
Péptidos , Proteómica , Proteómica/métodos , Péptidos/química , Proteínas/química , Concentración de Iones de Hidrógeno , Proteoma
2.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L287-L297, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29074491

RESUMEN

Statins, which are 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase competitive inhibitors, not only lower blood cholesterol but also exert pleiotropic and beneficial effects in various diseases. However, the effects of statins on acute lung injury (ALI) induced by hyperbaric oxygen (HBO) have not been investigated. The present study is the first to investigate the effects of simvastatin in ALI induced by HBO in 8- to 9-wk-old C57BL/6 mice exposed to 0.23 MPa [=2.3 atmosphere absolute (ATA)] hyperoxia (≥95% O2) for 6 h. Mice were either given simvastatin (20 mg·kg·-1·day-1) in saline or a saline vehicle for 3 days before oxygen exposure. Lung tissue, serum, and bronchoalveolar lavage fluid (BALF) were collected for analysis of proapoptotic proteins, low-density lipoprotein cholesterol (LDL-C) levels, and lung inflammation. Simvastatin treatment significantly reduced lung permeability, serum LDL-C levels, tissue apoptosis, and inflammation. However, simvastatin treatment had no effect on antioxidant enzyme activity, nicotinamide adenine dinucleotide phosphate oxidase 4 (NADPH4) expression, and Akt phosphorylation levels. Furthermore, we investigated the role of endothelial nitric oxide synthase (eNOS) in simvastatin protection through inhibiting eNOS activity with NG-nitro-l-arginine methyl ester (l-NAME; 20 mg/kg). Results showed that the beneficial effects of simvastatin on ALI induced by HBO (antiinflammatory, antiapoptotic, lipid lowering, and reduction in lung permeability) were reversed. These results showed that simvastatin curbs HBO-induced lung edema, permeability, inflammation, and apoptosis via upregulating eNOS expression and that simvastatin could be an effective therapy to treat prolonged HBO exposure.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Anticolesterolemiantes/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Oxigenoterapia Hiperbárica/efectos adversos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Simvastatina/farmacología , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/etiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Activación Transcripcional
3.
Undersea Hyperb Med ; 45(3): 351-362, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30028921

RESUMEN

Nuclear factor kappa B (NF-κB) is the critical transcriptional factor in the pathogenesis of acute lung injury (ALI). NF-κB regulates the expression changes of inflammatory factors such as tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin 6 (IL-6). In a previous study we showed that decompression sickness (DCS) caused by simulated unsafe fast buoyancy ascent escape (FBAE) could result in ALI, which was characterized by expression changes of inflammatory factors in rat lung tissue. The purpose of the present work was to study the roles of NF-κB and TNF-α in the process of DCS-induced rat lung injury caused by simulated unsafe FBAE. The research methods aimed to detect the rat lung tissue messenger ribonucleic acid (mRNA) and protein level variations of NF-κB, inhibitory ×B (I×B), TNF-α, IL-1ß, IL-6, IL-10 and IL-13 by using pretreatment of the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and TNF-α antibody (Ab). Our experimental results demonstrated that PDTC could improve the survival rate of the rats with DCS caused by unsafe FBAE more effectively than TNF-α Ab. However, the inhibition of TNF-α Ab on the nuclear translocated protein expression of NF-κB was more effective than PDTC. Both PDTC and TNF-α Ab can abrogate the increment of the rat lung tissue mRNA levels of TNF-α, IL-1ß, IL-6 and protein levels of NF-κB, TNF-α, IL-1ß effectively and increase the rat lung tissue content of I×B significantly. In conclusion, TNF-α-mediated NF-κB signaling may be one of the critical signaling pathways in the pathogenesis of DCS-induced rat lung injury caused by simulated unsafe FBAE. PDTC may ameliorate this type of injury partly through inhibiting the NF-κB pathway.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Antioxidantes/farmacología , Enfermedad de Descompresión/complicaciones , Interleucinas/metabolismo , FN-kappa B/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , FN-kappa B/antagonistas & inhibidores , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
4.
Hepatology ; 60(1): 349-61, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24711261

RESUMEN

UNLABELLED: A better understanding of hepatocyte senescence could be used to treat age-dependent disease processes of the liver. Whether continuously proliferating hepatocytes could avoid or reverse senescence has not yet been fully elucidated. We confirmed that the livers of aged mice accumulated senescent and polyploid hepatocytes, which is associated with accumulation of DNA damage and activation of p53-p21 and p16(ink4a)-pRB pathways. Induction of multiple rounds continuous cell division is hard to apply in any animal model. Taking advantage of serial hepatocyte transplantation assays in the fumarylacetoacetate hydrolase-deficient (Fah(-/-)) mouse, we studied the senescence of hepatocytes that had undergone continuous cell proliferation over a long time period, up to 12 rounds of serial transplantations. We demonstrated that the continuously proliferating hepatocytes avoided senescence and always maintained a youthful state. The reactivation of telomerase in hepatocytes after serial transplantation correlated with reversal of senescence. Moreover, senescent hepatocytes harvested from aged mice became rejuvenated upon serial transplantation, with full restoration of proliferative capacity. The same findings were also true for human hepatocytes. After serial transplantation, the high initial proportion of octoploid hepatocytes decreased to match the low level of youthful liver. CONCLUSION: These findings suggest that the hepatocyte "ploidy conveyer" is regulated differently during aging and regeneration. The findings of reversal of hepatocyte senescence could enable future studies on liver aging and cell therapy.


Asunto(s)
Proliferación Celular , Senescencia Celular/fisiología , Hepatocitos/citología , Hepatocitos/trasplante , Regeneración Hepática/fisiología , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citometría de Flujo , Hepatocitos/fisiología , Hidrolasas/genética , Operón Lac , Hígado/citología , Hígado/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Poliploidía , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
Undersea Hyperb Med ; 42(1): 15-22, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094300

RESUMEN

Fast buoyancy ascent escape is one of the major naval submarine escape maneuvers. Decompression sickness (DCS) is the major bottleneck to increase the depth of fast buoyancy ascent escape. Rapid decompression induces the release of inflammatory mediators and results in tissue inflammation cascades and a protective anti-inflammatory response. In our previous study, we found that DCS caused by simulated fast buoyancy ascent escape could induce acute lung injury (ALI) and the expression changes of the proinflammatory cytokines: tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß and IL-6 in rat lung tissue. In order to study the expression change characteristics of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 in the rat lung of DCS caused by simulated fast buoyancy ascent escape, we detected the rat lung mRNA and protein levels of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 at 0.5 hour after DCS caused by simulated fast buoyancy ascent escape (fast escape group), compared with the normal control group (control group) and diving DCS (decompression group). We observed that DCS caused by simulated fast buoyancy ascent escape could increase the mRNA levels of TNF-α, IL-1ß, IL-6, IL-10, and the protein levels of TNF-α, IL-10 in rat lung tissue. At the same time, we found that the protein level of IL-13 was also downregulated in rat lung tissue. TNF-α, IL-10 and IL-13 may be involved in the process of the rat lung injury of DCS caused by simulated fast buoyancy ascent escape. In conclusion, the expression changes of inflammatory factors in the rat lung of DCS caused by simulated fast buoyancy ascent escape were probably different from that in the rat lung of diving DCS, which indicated that the pathological mechanism of DCS caused by simulated fast buoyancy ascent escape might be different from that of diving DCS.


Asunto(s)
Enfermedad de Descompresión/metabolismo , Interleucinas/metabolismo , Pulmón/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Enfermedad de Descompresión/etiología , Enfermedad de Descompresión/mortalidad , Enfermedad de Descompresión/patología , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucinas/genética , Pulmón/patología , Masculino , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Medicina Submarina , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
6.
Exp Lung Res ; 40(3): 105-16, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24624894

RESUMEN

Recent studies have demonstrated that peroxisome proliferator-activated receptor-beta/delta (PPAR-ß/δ) has a protective effect during lung injury induced by bleomycin and polymicrobial sepsis, but its function in pulmonary oxygen toxicity is unknown. In this study, we used GW0742, a PPAR-ß/δ agonist, and GSK0660, a PPAR-ß/δ antagonist, to test the role of PPAR-ß/δ in lung injury due to hyperbaric oxygen (HBO2) exposure. Lung injury was induced in rats by HBO2 exposure (2.3 ATA, 100%O2, 8 hours). Sixty male Sprague-Dawley rats were randomly divided into 6 groups: air+vehicle, air+GW0742, air+GSK0660, HBO2+vehicle, HBO2+GW0742, and HBO2+GSK0660. Rats were injected with vehicle or GW0742 (0.3 mg/kg, i.p.) or GSK0660 (1 mg/kg, i.p.) at 1 hour, 6 hours, and 12 hours before either air or oxygen exposure. Administration of GW0742 to rats exposed to HBO2 significantly reduced the observed lung injury, extravascular lung water, total protein levels in bronchoalveolar lavage fluid, and the levels of iNOS and nNOS in the lungs when compared to untreated rats exposed to HBO2. Treatment of rats with GSK0660 exacerbated lung injury and elevated the levels of nNOS and eNOS in the lungs. In addition, nNOS and eNOS knock-out mice were examined. The results indicated that after HBO2 exposure, the lung injury was obviously decreased in the nNOS(-/-)+GSK0660 mice compared to the wild-type +GSK0660 mice; furthermore, administration of GSK0660 significantly elevated the lung injury in the eNOS(-/-) mice. Collectively, these data indicate that PPAR-ß/δ activation can protect against pulmonary oxygen toxicity in the lungs of rats through changes in the expression of NOS.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Óxido Nítrico Sintasa/metabolismo , Oxígeno/efectos adversos , PPAR delta/metabolismo , PPAR-beta/metabolismo , Lesión Pulmonar Aguda/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , PPAR-beta/agonistas , PPAR-beta/antagonistas & inhibidores , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Sulfonas , Tiazoles , Tiofenos , Regulación hacia Arriba
7.
Undersea Hyperb Med ; 41(1): 1-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24649711

RESUMEN

OBJECTIVE: To observe MMP9 expression in rat lungs under different degrees of hyperbaric oxygen (HBO2) exposure and to observe the relationship of tissue damage and apoptosis rate in the lung tissue. METHODS: 40 Sprague Dawley (SD) rats were randomly divided into five groups: 250 kPa oxygen exposure for two-, four-, six- and eight-hour exposures and a "normal group," each n = 8. After hyperbaric oxygen treatment, the rats were euthanized immediately to collect lung tissue. We used HE staining to detect the pathological changes and immunohistochemistry to detect in situ expression of matrix metalloproteinase 9 (MMP9). Then we tested the expression level of caspase 3 in lung tissue by Western Blot. To understand the antioxidant capacity changes, we detected superoxide dismutase (SOD) activities and malondialdehyde (MDA) contents in lung tissue during HBO2 exposure. Finally, we exposed MMP9 knockout or wild-type mice under hyperbaric oxygen for six hours, and detected the pathological changes with HE staining. RESULT: Lung tissue damage changed gradually under different degrees of hyperbaric oxygen exposure, but eased in the six-hour group. However, MMP9 expression decreased initially and then was upregulated until it reached the peak after six hours of exposure; it then reduced significantly after an eight-hour exposure. Active caspase 3 reached the highest level after eight hours. While SOD activity was upregulated only after six hours of HBO2 exposure, MDA content increased after eight hours of exposure. CONCLUSIONS: MMP9 expression was elevated after exposure to hyperbaric oxygen. This is a compensatory mechanism of the body's antioxidant response by regulating the inflammatory response. This in turn helps to reduce the apoptosis rate and protects lung tissue from oxygen toxicity.


Asunto(s)
Apoptosis , Oxigenoterapia Hiperbárica , Pulmón/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Caspasa 3/metabolismo , Técnicas de Inactivación de Genes , Pulmón/metabolismo , Pulmón/patología , Masculino , Malondialdehído/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Regulación hacia Arriba
8.
Comput Biol Med ; 174: 108457, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38599071

RESUMEN

Glioma is a common malignant brain tumor with great heterogeneity and huge difference in clinical outcomes. Although lymphotoxin (LT) beta receptor (LTBR) has been linked to immune system and response development for decades, the expression and function in glioma have not been investigated. To confirm the expression profile of LTBR, integrated RNA-seq data from glioma and normal brain tissues were analyzed. Functional enrichment analysis, TMEscore analysis, immune infiltration, the correlation of LTBR with immune checkpoints and ferroptosis, and scRNAseq data analysis in gliomas were in turn performed, which pointed out that LTBR was pertinent to immune functions of macrophages in gliomas. In addition, after being trained and validated in the tissue samples of the integrated dataset, an LTBR DNA methylation-based prediction model succeeded to distinguish gliomas from non-gliomas, as well as the grades of glioma. Moreover, by virtue of the candidate LTBR CpG sites, a prognostic risk-score model was finally constructed to guide the chemotherapy, radiotherapy, and immunotherapy for glioma patients. Taken together, LTBR is closely correlated with immune functions in gliomas, and LTBR DNA methylation could serve as a biomarker for diagnosis and prognosis of gliomas.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas , Glioma , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Metilación de ADN/genética , Glioma/inmunología , Glioma/genética , Glioma/metabolismo
9.
Cancer Med ; 12(24): 22170-22184, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38093622

RESUMEN

OBJECTIVE: As a single-transmembrane protein of the FXYD family, FXYD6 plays different roles under physiological and pathological status, especially in the nervous system. This study aims to identify FXYD6 as a biomarker for glioma, by analyzing its expression and methylation patterns. METHODS: Using TCGA and GTEx datasets, we analyzed FXYD6 expression in various tissues, confirming its levels in normal brain and different glioma grades via immunoblotting and immunostaining. FXYD6 biological functions were explored through enrichment analysis, and tumor immune infiltration was assessed using ESTIMATE and TIMER algorithms. Pearson correlation analysis probed FXYD6 associations with biological function-related genes. A glioma detection model was developed using FXYD6 methylation data from TCGA and GEO. Consistently, a FXYD6 methylation-based prognostic model was constructed for glioma via LASSO Cox regression. RESULTS: FXYD6 was observed to be downregulated in GBM and implicated in a range of cellular functions, including synapse formation, cell junctions, immune checkpoint, ferroptosis, EMT, and pyroptosis. Hypermethylation of specific FXYD6 CpG sites in gliomas was identified, which could be used to build a diagnostic model. Additionally, FXYD6 methylation-based prognostic model could serve as an independent factor as well. CONCLUSIONS: FXYD6 is a promising biomarker for the diagnosis and prognosis of glioma, with its methylation-based prognostic model serving as an independent factor. This highlights its potential in clinical application for glioma management.


Asunto(s)
Metilación de ADN , Glioma , Humanos , Biomarcadores , Glioma/diagnóstico , Glioma/genética , Algoritmos , Encéfalo , Pronóstico , Canales Iónicos
10.
Transfus Apher Sci ; 47(3): 305-11, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22947334

RESUMEN

Many biological molecules in the stored blood were involved in transfusion-related immunomodulation. One important effect was the differentiation bias of immune cells to Th2 type. In this study, we observed the immune regulation of extracellular ubiquitin accummulated in the plasma of whole blood on the differentiation of T help cells in vitro using ELISA and quantitative fluorescence real-time PCR with the help of LPS stimulated human peripheral blood mononuclear cells. We found extracellular ubiquitin promoted Th2 cytokine IL-4 production and Th2-inducing transcription factor STAT6 expression, but inhibited Th1 cytokine IFN-γ and Th1-inducing transcription factor T-bet, at the same time, proinflammation cytokine TNF-α was also inhibited. These findings probably contribute to the potent evidence that extracellular ubiquitin plays an indispensable role in the differentiation of T help cells which is crucial to the effects of transfusion-related immunomodulation.


Asunto(s)
Citocinas/biosíntesis , ARN Mensajero/sangre , Células TH1/metabolismo , Células Th2/metabolismo , Factores de Transcripción/sangre , Ubiquitina/sangre , Recolección de Muestras de Sangre , Transfusión Sanguínea/métodos , Citocinas/genética , Citocinas/metabolismo , Humanos , ARN Mensajero/biosíntesis
11.
Cell Death Dis ; 13(2): 176, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210426

RESUMEN

Microvascular invasion (MVI) is presently evaluated as a high-risk factor to be directly relative to postoperative prognosis of hepatocellular carcinoma (HCC). Up to now, diagnosis of MVI mainly depends on the postoperative pathological analyses with H&E staining assay, based on numbers and distribution characteristics of MVI to classify the risk levels of MVI. However, such pathological analyses lack the specificity to discriminate MVI in HCC specimens, especially in complicated pathological tissues. In addition, the efficiency to precisely define stages of MVI is not satisfied. Thus, any biomarker for both conforming diagnosis of MVI and staging its levels will efficiently and effectively promote the prediction of early postoperative recurrence and metastasis for HCC. Through bioinformatics analysis and clinical sample verification, we discovered that Stathmin 1 (STMN1) gene was significantly up-regulated at the locations of MVI. Combining STMN1 immunostaining with classic H&E staining assays, we established a new protocol for MVI pathological diagnosis. Next, we found that the degrees of MVI risk could be graded according to expression levels of STMN1 for prognosis prediction on recurrence rates and overall survival in early HCC patients. STMN1 affected epithelial-mesenchymal transformation (EMT) of HCC cells by regulating the dynamic balance of microtubules through signaling of "STMN1-Microtubule-EMT" axis. Inhibition of STMN1 expression in HCC cells reduced their lung metastatic ability in recipients of mouse model, suggesting that STMN1 also could be a potential therapeutic target for inhibiting HCC metastasis. Therefore, we conclude that STMN1 has potentials for clinical applications as a biomarker for both pathological diagnosis and prognostic prediction, as well as a therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Estatmina , Animales , Biomarcadores , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Ratones , Invasividad Neoplásica , Estudios Retrospectivos , Estatmina/genética
12.
J Clin Invest ; 132(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35239512

RESUMEN

Y-box-binding protein 1 (YB-1) is a multifunctional RNA binding protein involved in virtually every step of RNA metabolism. However, the functions and mechanisms of YB-1 in one of the most aggressive cancers, glioblastoma, are not well understood. In this study, we found that YB-1 protein was markedly overexpressed in glioblastoma and acted as a critical activator of both mTORC1 and mTORC2 signaling. Mechanistically, YB-1 bound the 5'UTR of CCT4 mRNA to promote the translation of CCT4, a component of the CCT chaperone complex, that in turn activated the mTOR signaling pathway by promoting mLST8 folding. In addition, YB-1 autoregulated its own translation by binding to its 5'UTR, leading to sustained activation of mTOR signaling. In patients with glioblastoma, high protein expression of YB-1 correlated with increased expression of CCT4 and mLST8 and activated mTOR signaling. Importantly, the administration of RNA decoys specifically targeting YB-1 in a mouse xenograft model resulted in slower tumor growth and better survival. Taken together, these findings uncover a disrupted proteostasis pathway involving a YB-1/CCT4/mLST8/mTOR axis in promoting glioblastoma growth, suggesting that YB-1 is a potential therapeutic target for the treatment of glioblastoma.


Asunto(s)
Glioblastoma , Proteína 1 de Unión a la Caja Y , Regiones no Traducidas 5' , Animales , Línea Celular Tumoral , Chaperonina con TCP-1 , Glioblastoma/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
13.
J Cell Biochem ; 106(1): 16-24, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19021148

RESUMEN

BMI-1 (B-cell-specific Moloney murine leukemia virus integration site 1), a novel oncogene, has attracted much attention in recent years for its involvement in the initiation of a variety of tumors. Recent evidence showed that BMI-1 was highly expressed in neoplastic skin lesions. However, whether dysregulated BMI-1 expression is causal for the transformation of skin cells remains unknown. In this study, we stably expressed BMI-1 in a human keratinocyte cell line, HaCaT. The expression of wild-type BMI-1 induced the malignant transformation of HaCaT cells in vitro. More importantly, we found that expression of BMI-1 promoted formation of squamous cell carcinomas in vivo. Furthermore, we showed that BMI-1 expression led to the downregulation of tumor suppressors, such as p16INK4a and p14ARF, cell adhesion molecules, such as E-Cadherin, and differentiation related factor, such as KRT6. Therefore, our findings demonstrated that dysregulated BMI-1 could indeed lead to keratinocytes transformation and tumorigenesis, potentially through promoting cell cycle progression and increasing cell mobility.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Queratinocitos/patología , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones SCID , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
14.
Mol Med Rep ; 19(2): 1272-1283, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30569161

RESUMEN

Apolipoprotein M (ApoM) is a type of apolipoprotein. It is well known that high­density lipoprotein (HDL) decreases inflammatory responses via the apoM­sphingosine­1­phosphate (S1P) pathway. The present study further investigated the importance of ApoM in the inhibitory effects of HDL on inflammation. Mice with an apoM gene deficiency (apoM­/­) were employed to investigate the effects of ApoM on the expression of interleukin­1ß (IL­1ß), monocyte chemotactic protein­1 (MCP­1), S1P receptor­1 (S1PR1) and 3ß­hydroxysterol Δ­24­reductase (DHCR24), as compared with in wild­type mice (apoM+/+). Furthermore, cell culture experiments were performed using a permanent human hybrid endothelial cell line (EA.hy926). Cells were cultured in the presence of recombinant human apoM (rec­apoM) or were induced to overexpress apoM (apoMTg); subsequently, cells were treated with tumor necrosis factor­α (TNF­α), in order to investigate the effects of ApoM on IL­1ß and MCP­1. The results demonstrated that the mRNA expression levels of IL­1ß and MCP­1 were significantly higher in the liver following administration of lipopolysaccharide in apoM­/­ mice compared with in apoM+/+ mice. In cell culture experiments, when cells were pre­cultured with rec­apoM or were engineered to overexpress apoM (apoMTg), they exhibited decreased expression levels of IL­1ß and MCP­1 following TNF­α treatment compared with in normal apoM­expressing cells (apoMTgN). Furthermore, the mRNA expression levels of IL­1ß and MCP­1 were significantly elevated following addition of the S1PR1 inhibitor W146, but not by the scavenger receptor class B type I inhibitor, block lipid transport­1 (BLT­1), in apoMTg cells prior to TNF­α treatment. Conversely, there were no differences in these inflammatory biomarkers under the same conditions in apoMTgN cells. The mRNA expression levels of DHCR24 were significantly reduced by the addition of BLT­1 prior to TNF­α treatment in apoMTg cells; however, there was no difference in the expression of this inflammatory biomarker in apoMTgN cells. In conclusion, ApoM displayed inhibitory effects against the inflammatory response in vivo and in vitro; these effects may be induced via the S1PR1 and DHCR24 pathways.


Asunto(s)
Apolipoproteínas M/metabolismo , Inflamación/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/fisiología , Animales , Biomarcadores/metabolismo , Línea Celular , Quimiocina CCL2/metabolismo , Humanos , Lipoproteínas HDL/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Esfingosina-1-Fosfato , Factor de Necrosis Tumoral alfa/metabolismo
15.
Lung Cancer ; 123: 70-75, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30089598

RESUMEN

OBJECTIVES: The prognostic value of TP53 in advanced non-small-cell lung cancer (NSCLC) is unclear. Whether different mutated exon has different prognostic value is unknown. We sought to reveal the prognostic value of TP53 in advanced NSCLC, as well as the correlation with EGFR mutation. MATERIALS AND METHODS: Information regarding TP53 and EGFR alterations and patients' survival time in advanced NSCLC was downloaded from the Cancer Genome Atlas Database. We further subdivided TP53 and EGFR mutation into subgroups based on different mutation exon, and then evaluated the distribution of different mutation exon as well as the prognostic value. RESULTS AND CONCLUSION: Overall, 1441 pieces of data from 1441 metastatic NSCLC patient were collected. Mutation rate of TP53 was 56.1% (809/1441). TP53 mutation was a negative prognostic factor for OS. The estimated survival time for wild type TP53 and mutated TP53 was 27.0 months (95% CI, not reached) and 19 months (95% CI, 16.62 to 21.38), respectively, (p < 0.001). We divided TP53 mutations into 4 groups, OS in these 4 groups was 27 months (95% CI, not reached), not reached, 21 months (95% CI, 17.16 to 24.84) and 13 months (95% CI, 10.39 to 15.61). The difference was statistically significant (p < 0.001). Patients with EGFR exon 19/21 or non-exon 19/21 mutation demonstrated a higher rate of mutated type TP53 than EGFR wild type patients. Survival curve in EGFR wild type patients indicated that TP53 wild type patients had the best prognosis. In patients with exon 19/21 mutated EGFR, the trend was the same (P < 0.001).TP53 mutation is a negative prognostic factor in advanced NSCLC, different mutated exon has different prognostic value. When coupled with EGFR mutation, we can predict the prognosis of advanced NSCLC patients more accurately.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Mutación , Proteína p53 Supresora de Tumor/genética , Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas/patología , Receptores ErbB/genética , Exones , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Tasa de Mutación , Estadificación de Neoplasias , Pronóstico
16.
Cell Death Dis ; 9(5): 575, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29760381

RESUMEN

It is well known that induction of hepatocyte senescence could inhibit the development of hepatocellular carcinoma (HCC). Until now, it is still unclear how the degree of liver injury dictates hepatocyte senescence and carcinogenesis. In this study, we investigated whether the severity of injury determines cell fate decisions between hepatocyte senescence and carcinogenesis. After testing of different degrees of liver injury, we found that hepatocyte senescence is strongly induced in the setting of severe acute liver injury. Longer-term, moderate liver injury, on the contrary did not result into hepatocyte senescence, but led to a significant incidence of HCC instead. In addition, carcinogenesis was significantly reduced by the induction of severe acute injury after chronic moderate liver injury. Meanwhile, immune surveillance, especially the activations of macrophages, was activated after re-induction of senescence by severe acute liver injury. We conclude that severe acute liver injury leads to hepatocyte senescence along with activating immune surveillance and a low incidence of HCC, whereas chronic moderate injury allows hepatocytes to proliferate rather than to enter into senescence, and correlates with a high incidence of HCC. This study improves our understanding in hepatocyte cell fate decisions and suggests a potential clinical strategy to induce senescence to treat HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Senescencia Celular , Hepatocitos/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/lesiones , Hígado/metabolismo , Enfermedad Aguda , Animales , Carcinoma Hepatocelular/patología , Hepatocitos/patología , Hígado/patología , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados
17.
Front Biosci ; 12: 1691-8, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17127414

RESUMEN

The liver regenerates by progenitor cells when it is damaged in chronic liver diseases and extensive damage. The progenitor cells, also termed "oval cells" according to their morphological traits, can differentiate into hepatocytes and bile duct cells in vivo. To better understand the transcriptional pattern that accompanies the hepatic differentiation of oval cells, we applied cDNA microarray to analyze the oval cell-derived liver epithelial progenitor cells (LEPCs) during in vitro induced differentiation. Upon exposure to sodium butyrate, a histone deacetylase inhibitor, cultured LEPCs differentiate and express functional hepatocyte markers albumin, tryptophan 2, 3-dioxygenase and alcohol dehydrogenase. For expression profiling, cells were harvested at 6 h, 12 h, 1 d, 3 d and 7 d after exposure to sodium butyrate. After analyzing the microarray data by SOM clustering, total of 796 differentially regulated genes were grouped into 48 clusters. Consistent with the phenotype change of LEPCs after sodium butyrate treatment, many hepatocyte functional genes are revealed by analyzing the clusters containing genes up-regulated through all the time points. The clusters, containing down-regulated genes immediately after the induction, are also analyzed. The microarray data was validated by analyzing the expression of selected genes by quantitative real-time PCR. A set of genes expressed synergistically in these clusters may play a central role during the process of differentiation. Sodium butyrate decreases cyclin B1 and Cdk4 expression, which would be associated with LEPCs growth arrest shortly after treatment. Bmi1, a polycomb group protein, is also down-regulated immediately after treatment and remains at a low level during the induction. These findings highlight the key molecular mechanisms by which sodium butyrate, mediates its effects on cell growth arrest and induction of differentiation. In conclusion, our data reflect a global view of gene expression during hepatic differentiation of LEPCs induced by sodium butyrate.


Asunto(s)
Hepatocitos/metabolismo , Hígado/citología , Células Madre/metabolismo , Transcripción Genética , Animales , Butiratos/farmacología , Diferenciación Celular , Línea Celular , Ciclina B/genética , Ciclina B/metabolismo , Ciclina B1 , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Hepatocitos/citología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Células Madre/citología , Células Madre/efectos de los fármacos
18.
Artículo en Zh | MEDLINE | ID: mdl-26827528

RESUMEN

OBJECTIVE: To investigate the effect of different pressure oxygen pre-breathing in preventing decompression sickness of rats. METHODS: Forty male SD rats were randomly divided into 4 groups: decompression sickness (DCS) group and three oxygen pre-breathing groups with 1 ATA, 2 ATA and 3 ATA pressure respectively. The rats of DCS group were placed in the hyperbaric chamber and the chamber was compressed evenly within 3 minutes to depths of 7 absolute atmosphere(ATA) and held at the designated depth for 60 min, then decompressed (3 min) at constant speed to the surface pressure. After that, the rats were taken out for further detection. While the rats of oxygen pretreatment groups pre-breathed different pressure oxygen for 20 min before entering into chamber. The mortality and behavioral of rats were observed with 30 min post decompression. The dry/wet ratio of the lung, protein levels in the bronchoalveolar lavage fluid (BALF), and the inflammatory cytokine tumor necrosis factor (TNF-alpha) expression were also tested. RESULTS: Compared with that of the DCS group, the mortality and morbidity of oxygen pre-breathe groups didn't change obviously. But the total BALF protein level and the inflammatory cytokine TNF-alpha expression of 1 ATA oxygen pre-breathe group were obviously decreased, while the dry/wet ratio of lung as obviously increased instead (P < 0.05). CONCLUSION: Although preoxygenation can' t obviously change the mortality and mobidity of rats, normal pressure oxygen pre-breathing can mitigate the protein infiltration in BALF and the expression of inflammatory cytokine in lung tissue.


Asunto(s)
Enfermedad de Descompresión , Oxígeno/fisiología , Animales , Líquido del Lavado Bronquioalveolar/química , Buceo , Pulmón/patología , Presión , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
19.
Respir Physiol Neurobiol ; 211: 9-16, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25784626

RESUMEN

Inflammation and platelet activation are critical phenomena in the setting of decompression sickness. Clopidogrel (Clo) inhibits platelet activation and may also reduce inflammation. The goal of this study was to investigate if Clo had a protective role in decompression sickness (DCS) through anti-inflammation way. Male Sprague-Dawley rats (n=111) were assigned to three groups: control+vehicle group, DCS+vehicle, DCS+Clo group. The experimental group received 50 mg/kg of Clo or vehicle for 3 days, then compressed to 1,600 kPa (150 msw) in 28 s, maintained at 150 msw for 242 s and decompressed to surface at 3m/s. In a control experiment, rats were also treated with vehicle for 3 days and maintained at atmospheric pressure for an equivalent period of time. Clinical assessment took place over a period of 30 min after surfacing. At the end, blood samples were collected for blood cells counts and cytokine detection. The pathology and the wet/dry ratio of lung tissues, immunohistochemical detection of lung tissue CD41 expression, the numbers of P-selectin positive platelets and platelet-leukocyte conjugates in blood were tested. We found that Clo significantly reduced the DCS mortality risk (mortality rate: 11/45 with Clo vs. 28/46 in the untreated group, P<0.01). Clo reduced the lung injury, the wet/dry ratio of lung, the accumulation of platelet and leukocyte in lung, the fall in platelet count, the WBC count, the numbers of activated platelets and platelet-leukocyte complexes in peripheral blood. It was concluded that Clo can play a protective role in decompression sickness through reducing post-decompression platelet activation and inflammatory process.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Enfermedad de Descompresión/tratamiento farmacológico , Enfermedad de Descompresión/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ticlopidina/análogos & derivados , Animales , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Clopidogrel , Citocinas/metabolismo , Enfermedad de Descompresión/sangre , Enfermedad de Descompresión/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Pulmón/patología , Masculino , Tamaño de los Órganos , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Presión , Ratas Sprague-Dawley , Ticlopidina/farmacología , Resultado del Tratamiento
20.
Respir Physiol Neurobiol ; 199: 9-18, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24780550

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)-ß/δ is a transcription factor that belongs to the PPAR family, but the role of PPAR-ß/δ in acute lung injury (ALI) induced by hyperbaric oxygen is unknown. In this study we investigated if PPAR-ß/δ activation protects from hyperoxia-induced ALI in a rat model. ALI was induced by prolonged hyperbaric oxygen (HBO2) (2.3ATA, 100% O2) for 8h. Administration of PPAR-ß/δ agonist GW0742 (0.3mg/kg, i.p.) at 1 and 6h prior to HBO2 exposure significantly reduced the (1) lung injury, (2) proinflammatory cytokines (TNF-α, IL-1ß, IL-6), (3) apoptosis (Bax/Bcl-2, cleaved-caspase-3 and TUNEL), (4) nuclear factor (NF)-κB expression level and DNA binding activity in the nucleus, and (5) extracellular signal-regulated kinase (ERK)1/2 phosphorylation and markedly elevated (6) superoxide dismutase and glutathione peroxidase activities as well as (7) IκB expression. However, administration of the PPAR-ß/δ antagonist GSK0660 abolished these protective effects. These findings indicate that activation of PPAR-ß/δ ameliorates hyperoxia-induced ALI in rats by up-regulating antioxidant enzyme activity as well as suppressing inflammation and apoptosis.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/fisiopatología , Hiperoxia/complicaciones , PPAR delta/metabolismo , PPAR-beta/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , PPAR-beta/agonistas , PPAR-beta/antagonistas & inhibidores , Presión , Distribución Aleatoria , Ratas Sprague-Dawley , Fármacos del Sistema Respiratorio/farmacología , Sulfonas/farmacología , Tiazoles/farmacología , Tiofenos/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA