Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cardiothorac Vasc Anesth ; 38(7): 1569-1576, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38594156

RESUMEN

Patient blood management (PBM) guidelines for patients undergoing cardiac surgery under cardiopulmonary bypass (CPB) have increased during the past decade, and pharmacotherapy plays an important role in PBM. In the face of the undefined consistency in the methodologic quality and pharmacotherapy recommendations across multiple guidelines, this study exclusively evaluated methodologies of the related guideline development process, and compiled medication recommendations of PBM for cardiac surgery patients. PBM guidelines for cardiac surgery under CPB were searched through some mainstream literature and guideline databases from database establishment to May 15, 2023. Nine guidelines meeting inclusion criteria were included in this study. The quality of the guidelines was evaluated using the Appraisal of Guidelines for Research and Evaluation II (AGREE II) tool. "Stakeholder involvement" received the lowest mean score of 49.38% in the AGREE II scoring among the guidelines. PBM for cardiac surgery patients spans the perioperative phase. Drug therapy strategies of PBM for cardiac surgery patients involve anemia therapy, perioperative administration of antithrombotic drugs, intraoperative anticoagulation, and the use of hemostatic drugs. Unlike for adults, there is less evidence about the management of antithrombotic drugs and hemostatic drugs for pediatric cardiac surgery patients. Recombinant activated factor VII (rFVIIa) and desmopressin (DDAVP) are not recommended after pediatric cardiac surgery, whereas prothrombin complex concentrate could be considered in clinical trials. As for the controversies regarding the administration of rFVIIa and DDAVP after adult cardiac surgery by different societies, clinicians should exercise their clinical judgment based on individual patient features.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Puente Cardiopulmonar , Guías de Práctica Clínica como Asunto , Humanos , Puente Cardiopulmonar/métodos , Puente Cardiopulmonar/normas , Procedimientos Quirúrgicos Cardíacos/métodos , Guías de Práctica Clínica como Asunto/normas
2.
Biochem Biophys Res Commun ; 642: 1-10, 2023 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-36535215

RESUMEN

BLOC1S1 is a common component of BLOC and BORC multiprotein complexes which play distinct roles in endosome and lysosome biology. Recent human mutations in BLOC1S1 associate with juvenile leukodystrophy. As leukodystrophy is linked to perturbed lysosomal lipid storage we explored whether BLOC1S1 itself modulates this biology. Given the central role of the liver in lipid storage, our investigations were performed in hepatocyte specific liver bloc1s1 knockout (LKO) mice and in human hepatocyte-like lines (HLCs) derived from inducible pluripotential stem cells (iPSCs) from a juvenile leukodystrophy subject's with bloc1s1 mutations and from isogenic corrected iPSCs. Here we show that hepatocyte lipid stores are diminished in parallel with increased lysosomal content, increased lysosomal lipid uptake and lipolysis in LKO mice. The lysosomal lipolysis program was independent of macro- and chaperone-mediated lipophagy but dependent on cellular lysosome content. In parallel, genetic induction of lysosomal biogenesis in a transformed hepatocyte cell line replicated depletion of intracellular lipid stores. Interestingly bloc1s1 mutant and isogenic corrected HLCs both showed normal lysosomal enzyme activity. However, relative to the isogenic corrected HLCs, mutant bloc1s1 HLCs showed reduced lysosomal content and increased lipid storage. Together these data show distinct phenotypes in human mutant HLCs compared to murine knockout cells. At the same time, human blcs1s1 mutation and murine hepatocyte bloc1s1 depletion disrupt lysosome content and the cellular lipid storage. These data support that BLOC1S1 modulates lysosome content and lipid handling independent of autophagy and show that lysosomal lipolysis is dependent on the cellular content of functional lysosomes.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Lipólisis , Animales , Ratones , Humanos , Hígado/metabolismo , Lisosomas/metabolismo , Factores de Transcripción/metabolismo , Trastornos del Metabolismo de los Lípidos/metabolismo , Autofagia , Lípidos , Proteínas del Tejido Nervioso/metabolismo
3.
J Cardiovasc Pharmacol ; 79(2): 192-198, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34117183

RESUMEN

ABSTRACT: Long noncoding RNA is one potential target for the treatment of various disorders. Here, we explored the role of Abhd11os in ischemia/reperfusion-induced myocardial injury, and preliminarily explored the regulatory mechanisms. Relative Abhd11os expression level was examined by qRT-PCR. Western blot was done to measure the expression of apoptotic-related proteins. Cell counting kit-8 assay and flow cytometry were performed to detect cell viability and apoptosis, respectively. ELISA assay was used to ensure the levels of lactate dehydrogenase, creatine kinase, and cardiac troponin I in serum. Besides, the infarct sizes were confirmed by 2,3,5-triphenyltetrazolium chloride and Evans blue staining. Apoptotic rate of cardiomyocytes in myocardial tissues was evaluated by TUNEL assay. Here, increased Abhd11os expression was found in rat myocardial ischemia/reperfusion injury (MIRI) model and hypoxia/reoxygenation-treated cardiomyocytes. Subsequently, our data in vitro showed that upregulation of Abhd11os inhibited proliferation of cardiomyocytes, but promoted cell apoptosis. In animal experiments, myocardial infarct size in MIRI rats was reduced by Abhd11os knockdown. Moreover, downregulation of Abhd11os inhibited apoptosis of cardiomyocytes. Overall, our results revealed that knockdown of Abhd11os could notably attenuate hypoxia/reoxygenation-induced myocardial injury through suppressing apoptosis of cardiomyocytes. These data suggest that Abhd11os may be a potential target for MIRI therapy.


Asunto(s)
Daño por Reperfusión Miocárdica , ARN Largo no Codificante , Animales , Apoptosis/genética , Hipoxia/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratas
4.
J Clin Lab Anal ; 36(11): e24723, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36189780

RESUMEN

BACKGROUND: Recently, a new type of programmed cell death, cuproptosis, has been identified to play important role in the progression of tumors. We constructed a cuproptosis-related long non-coding RNA (lncRNA) signature to predict the prognostic significance for head and neck squamous cell carcinoma (HNSCC). METHODS: The risk model was developed based on differentially expressed lncRNAs associated with cuproptosis. Principal component analysis was used to assess the validity. The Kaplan-Meier curves were analyzed to compare the overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS) values. The multivariate and univariate Cox regression analyses were used to evaluate the prognostic efficiency. Furthermore, the functional enrichment, immune cell infiltration, tumor mutation burden (TMB), and sensitivity toward chemotherapy were also explored. RESULTS: Six cuproptosis-related lncRNAs (AL109936.2, CDKN2A-DT, AC090587.1, KLF3-AS1, AL133395.1, and LINC01063) were identified to construct the independent prognostic predictor for HNSCC. The area under the curve and C-index values obtained using the risk model were higher than the values corresponding to the clinical factors. Analysis of Kaplan-Meier curves indicated that the OS, PFS, and DSS time recorded for the patients in the low-risk group were higher than the corresponding values recorded for the patients belonging to the high-risk group. By functional enrichment analysis, we observed that differentially expressed genes were enriched in the immune response and tumor-associated pathways. The patients characterized by a low-risk score exhibited better immune cell infiltration than the patients belonging to the other group. We also observed that the sensitivity of the individuals belonging to the low-risk group to chemotherapeutic agents (cisplatin, docetaxel, and paclitaxel) was higher than the sensitivity of those in the other group. CONCLUSIONS: A cuproptosis-related lncRNA-based signature that functioned as an independent prognosis predictor for HNSCC patients was constructed. The chemosensitivity of individual patients can be potentially predicted using this signature.


Asunto(s)
Apoptosis , Neoplasias de Cabeza y Cuello , ARN Largo no Codificante , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Pronóstico , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Cobre
5.
Hepatology ; 71(2): 643-657, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31344750

RESUMEN

BACKGROUND AND AIMS: The regenerative capacity of the liver plays a protective role against hepatotoxins and impaired regeneration exacerbates liver dysfunction in nonalcoholic fatty liver disease (NAFLD). Mitochondrial bioenergetic and -synthetic functions are important contributory factors in hepatic regeneration, and the control of mitochondrial protein acetylation is implicated in the mitochondrial susceptibility to liver stressors. Here, we evaluated the role of general control of amino acid synthesis 5 like 1 (GCN5L1), a mediator of mitochondrial metabolism and acetylation, in modulating murine liver regeneration (LR) in response to acute CCl4 -induced hepatotoxicity. APPROACH AND RESULTS: Initial metabolomic screening found that liver GCN5L1 knockout (LKO) mice have augmented glutaminolysis. Absence of GCN5L1 modified enzyme activity of liver-enriched glutaminase enzyme (glutaminase 2; GLS2), and GCN5L1 levels modulated GLS2 oligomerization and acetylation. This metabolic remodeling resulted in the elevation of α-ketoglutarate levels, which are known to activate mammalian target of rapamycin complex 1 (mTORC1). This signaling pathway was induced with increased phosphorylation of S6 kinase in LKO hepatocytes, and inhibition of glutaminolysis reversed aberrant mTORC1 signaling. At the same time, glutaminolysis, activity of GLS2, and activation of mTORC1 signaling were reversed by the genetic reintroduction of the mitochondrial isoform of GCN5L1 into LKO primary hepatocytes. Finally, LKO mice had a more robust regenerative capacity in response to CCl4 hepatoxicity, and this response was blunted by both the mTORC1 inhibitor, rapamycin, and by pharmacological blunting of glutaminolysis. CONCLUSIONS: These data point to a central role of glutaminolysis in modulating the regenerative capacity in the liver. Furthermore, inhibition of mitochondrial GCN5L1 to augment LR may be a useful strategy in disease states linked to hepatotoxicity.


Asunto(s)
Glutamina/metabolismo , Regeneración Hepática/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Proteínas Mitocondriales/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Masculino , Ratones , Transducción de Señal
6.
BMC Cardiovasc Disord ; 21(1): 300, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130651

RESUMEN

BACKGROUND: Despite significant progress in surgical treatment of hypoplastic left heart syndrome (HLHS), its mortality and morbidity are still high. Little is known about the molecular abnormalities of the syndrome. In this study, we aimed to probe into hub genes and key pathways in the progression of the syndrome. METHODS: Differentially expressed genes (DEGs) were identified in left ventricle (LV) or right ventricle (RV) tissues between HLHS and controls using the GSE77798 dataset. Then, weighted gene co-expression network analysis (WGCNA) was performed and key modules were constructed for HLHS. Based on the genes in the key modules, protein-protein interaction networks were conducted, and hub genes and key pathways were screened. Finally, the GSE23959 dataset was used to validate hub genes between HLHS and controls. RESULTS: We identified 88 and 41 DEGs in LV and RV tissues between HLHS and controls, respectively. DEGs in LV tissues of HLHS were distinctly involved in heart development, apoptotic signaling pathway and ECM receptor interaction. DEGs in RV tissues of HLHS were mainly enriched in BMP signaling pathway, regulation of cell development and regulation of blood pressure. A total of 16 co-expression network were constructed. Among them, black module (r = 0.79 and p value = 2e-04) and pink module (r = 0.84 and p value = 4e-05) had the most significant correlation with HLHS, indicating that the two modules could be the most relevant for HLHS progression. We identified five hub genes in the black module (including Fbn1, Itga8, Itga11, Itgb5 and Thbs2), and five hub genes (including Cblb, Ccl2, Edn1, Itgb3 and Map2k1) in the pink module for HLHS. Their abnormal expression was verified in the GSE23959 dataset. CONCLUSIONS: Our findings revealed hub genes and key pathways for HLHS through WGCNA, which could play key roles in the molecular mechanism of HLHS.


Asunto(s)
Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Síndrome del Corazón Izquierdo Hipoplásico/genética , ARN Mensajero/genética , Transcriptoma , Animales , Estudios de Casos y Controles , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Síndrome del Corazón Izquierdo Hipoplásico/diagnóstico por imagen , Síndrome del Corazón Izquierdo Hipoplásico/metabolismo , Ratones , Fenotipo , Mapas de Interacción de Proteínas , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal
7.
J Cell Sci ; 131(22)2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30333138

RESUMEN

Although GCN5L1 (also known as BLOC1S1) facilitates mitochondrial protein acetylation and controls endosomal-lysosomal trafficking, the mechanisms underpinning these disparate effects are unclear. As microtubule acetylation modulates endosome-lysosome trafficking, we reasoned that exploring the role of GCN5L1 in this biology may enhance our understanding of GCN5L1-mediated protein acetylation. We show that α-tubulin acetylation is reduced in GCN5L1-knockout hepatocytes and restored by GCN5L1 reconstitution. Furthermore, GCN5L1 binds to the α-tubulin acetyltransferase αTAT1, and GCN5L1-mediated α-tubulin acetylation is dependent on αTAT1. Given that cytosolic GCN5L1 has been identified as a component of numerous multiprotein complexes, we explored whether novel interacting partners contribute to this regulation. We identify RanBP2 as a novel interacting partner of GCN5L1 and αTAT1. Genetic silencing of RanBP2 phenocopies GCN5L1 depletion by reducing α-tubulin acetylation, and we find that RanBP2 possesses a tubulin-binding domain, which recruits GCN5L1 to α-tubulin. Finally, we find that genetic depletion of GCN5L1 promotes perinuclear lysosome accumulation and histone deacetylase inhibition partially restores lysosomal positioning. We conclude that the interactions of GCN5L1, RanBP2 and αTAT1 function in concert to control α-tubulin acetylation and may contribute towards the regulation of cellular lysosome positioning. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Acetiltransferasas/metabolismo , Hígado/metabolismo , Lisosomas/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Acetilación , Animales , Células HEK293 , Células HeLa , Hepatocitos/metabolismo , Humanos , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microtúbulos/metabolismo , Proteínas Mitocondriales , Cultivo Primario de Células , Transfección
8.
J Biol Chem ; 293(46): 17676-17684, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30323061

RESUMEN

Sirtuin 3 (SIRT3) deacetylates and activates several mitochondrial fatty acid oxidation enzymes in the liver. Here, we investigated whether the protein acetylase GCN5 general control of amino acid synthesis 5-like 1 (GCN5L1), previously shown to oppose SIRT3 activity, is involved in the regulation of hepatic fatty acid oxidation. We show that GCN5L1 abundance is significantly up-regulated in response to an acute high-fat diet (HFD). Transgenic GCN5L1 overexpression in the mouse liver increased protein acetylation levels, and proteomic detection of specific lysine residues identified numerous sites that are co-regulated by GCN5L1 and SIRT3. We analyzed several fatty acid oxidation proteins identified by the proteomic screen and found that hyperacetylation of hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit α (HADHA) correlates with increased GCN5L1 levels. Stable GCN5L1 knockdown in HepG2 cells reduced HADHA acetylation and increased activities of fatty acid oxidation enzymes. Mice with a liver-specific deletion of GCN5L1 were protected from hepatic lipid accumulation following a chronic HFD and did not exhibit hyperacetylation of HADHA compared with WT controls. Finally, we found that GCN5L1-knockout mice lack HADHA that is hyperacetylated at three specific lysine residues (Lys-350, Lys-383, and Lys-406) and that acetylation at these sites is significantly associated with increased HADHA activity. We conclude that GCN5L1-mediated regulation of mitochondrial protein acetylation plays a role in hepatic metabolic homeostasis.


Asunto(s)
Ácidos Grasos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Acetilación , Animales , Dieta Alta en Grasa/efectos adversos , Hígado Graso/prevención & control , Células Hep G2 , Humanos , Lisina/química , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Proteínas del Tejido Nervioso/genética , Oxidación-Reducción , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteómica , Sirtuina 3/genética
9.
J Neurosci ; 37(40): 9741-9758, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28883020

RESUMEN

As the population ages, an increasing number of people suffer from age-related cognitive impairment. However, the mechanisms underlying this process remain unclear. Here, we found that S-nitrosoglutathione reductase (GSNOR), the key enzyme that metabolizes intracellular nitric oxide (NO) and regulates S-nitrosation, was significantly increased in the hippocampus of both aging humans and mice. Transgenic mice overexpressing GSNOR exclusively in neurons showed cognitive impairment in behavioral tests, including the Morris water maze, fear conditioning, and the Y-maze test. We also found that GSNOR transgenic mice have LTP defects and lower dendrite spine density, whereas GSNOR knock-out mice rescued the age-related cognitive impairment. Analysis of S-nitrosation showed significantly decreased hippocampal CaMKIIα S-nitrosation in naturally aged mice and GSNOR transgenic mice. Consistent with the change in CaMKIIα S-nitrosation, the accumulation of CaMKIIα in the hippocampal synaptosomal fraction, as well as its downstream signaling targets p(S831)-GLUR1, was also significantly decreased. All these effects could be rescued in the GSNOR knock-out mice. We further verified that the S-nitrosation of CaMKIIα was responsible for the CaMKIIα synaptosomal accumulation by mutating CaMKIIα S-nitrosated sites (C280/C289). Upregulation of the NO signaling pathway rescued the cognitive impairment in GSNOR transgenic mice. In summary, our research demonstrates that GSNOR impairs cognitive function in aging and it could serve as a new potential target for the treatment of age-related cognitive impairment. In contrast to the free radical theory of aging, NO signaling deficiency may be the main mediator of age-related cognitive impairment.SIGNIFICANCE STATEMENT This study indicated that S-nitrosoglutathione reductase (GSNOR), a key protein S-nitrosation metabolic enzyme, is a new potential target in age-related cognitive impairment; and in contrast to the free radical theory of aging, NO signaling deficiency may be the main cause of this process. In addition, increased GSNOR expression during aging decreases S-nitrosation of CaMKIIα and reduces CaMKIIα synaptosomal accumulation. To our knowledge, it is for the first time to show the cellular function regulation of CaMKIIα by GSNOR-dependent S-nitrosation as a new post-translational modification after its phosphorylation was explored. These findings elucidate a novel mechanism of age-related cognitive impairment and may provide a new potential target and strategy for slowing down this process.


Asunto(s)
Envejecimiento/metabolismo , Alcohol Deshidrogenasa/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Trastornos del Conocimiento/metabolismo , Cognición/fisiología , Regulación Enzimológica de la Expresión Génica , Envejecimiento/genética , Alcohol Deshidrogenasa/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Trastornos del Conocimiento/genética , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Nitrosación/fisiología , Técnicas de Cultivo de Órganos
10.
Glia ; 66(11): 2427-2437, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30378174

RESUMEN

Loss of substantia nigra dopaminergic neurons results in Parkinson disease (PD). Degenerative PD usually presents in the seventh decade whereas genetic disorders, including mutations in PARK2, predispose to early onset PD. PARK2 encodes the parkin E3 ubiquitin ligase which confers pleotropic effects on mitochondrial and cellular fidelity and as a mediator of endoplasmic reticulum (ER) stress signaling. Although the majority of studies investigating ameliorative effects of parkin focus on dopaminergic neurons we found that astrocytes are enriched with parkin. Furthermore, astrocytes deficient in parkin display stress-induced elevation of nucleotide-oligomerization domain receptor 2 (NOD2), a cytosolic receptor integrating ER stress and inflammation. Given the neurotropic and immunomodulatory role of astrocytes we reasoned that parkin may regulate astrocyte ER stress and inflammation to control neuronal homeostasis. We show that, in response to ER stress, parkin knockdown astrocytes exhibit exaggerated ER stress, JNK activation and cytokine release, and reduced neurotropic factor expression. In coculture studied we demonstrate that dopaminergic SHSY5Y cells and primary neurons with the presence of parkin depleted astrocytes are more susceptible to ER stress and inflammation-induced apoptosis than wildtype astrocytes. Parkin interacted with, ubiquitylated and diminished NOD2 levels. Additionally, the genetic induction of parkin ameliorated inflammation in NOD2 expressing cells and knockdown of NOD2 in astrocytes suppressed inflammatory defects in parkin deficient astrocytes and concurrently blunted neuronal apoptosis. Collectively these data identify a role for parkin in modulating NOD2 as a regulatory node in astrocytic control of neuronal homeostasis.


Asunto(s)
Astrocitos/ultraestructura , Estrés del Retículo Endoplásmico/fisiología , Inflamación/patología , Factores de Crecimiento Nervioso/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , L-Lactato Deshidrogenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Crecimiento Nervioso/genética , Proteína Oncogénica p55(v-myc)/metabolismo , Oxidopamina/farmacología , Factor de Transcripción CHOP/metabolismo , Ubiquitina-Proteína Ligasas/genética
11.
Gene Expr ; 18(2): 135-147, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29409568

RESUMEN

Simultaneous mutations in CTNNB1 and activation of c-MET occur in 9%-12.5% of patients with hepatocellular carcinoma (HCC). Coexpression of c-MET-V5 and mutant ß-catenin-Myc in mouse liver by sleeping beauty transposon/transposase and hydrodynamic tail vein injection (SB-HTVI) led to the development of HCC with 70% molecular identity to the clinical subset. Using this model, we investigated the effect of EMD1214063, a highly selective c-MET inhibitor. Five weeks after SB-HTVI when tumors were established, EMD1214063 (10 mg/kg) was administered by gastric gavage as a single agent on 5-day-on/3-day-off schedule, compared to vehicle only control. Mice were harvested at 8 or 11 weeks posttreatment. Decreased p-MET, p-AKT, p-STAT3, and p-ERK proved in vivo efficacy of EMD1214063. We observed lower Ki-67, PCNA, V5-tag, and cyclin D1 after EMD1214063 treatment only at 8 weeks. Overall, no significant differences were observed in tumor burden between the groups, although EMD1214063 marginally but significantly improved overall survival by 1.5-2 weeks. Tumors remained α-fetoprotein+, did not show any differences in inflammation, and lacked fibrosis in either group. In conclusion, c-MET inhibition alone had a minor effect on Met-ß-catenin HCC at the early stages of HCC development. Thus, a single therapy with the c-MET inhibitor will be insufficient for sustained response in Met-ß-catenin HCC requiring assessment of additional combinations.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridazinas/uso terapéutico , Pirimidinas/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Masculino , Ratones , Mutación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridazinas/administración & dosificación , Piridazinas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , beta Catenina/genética
12.
J Card Surg ; 32(2): 151-153, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27990684

RESUMEN

OBJECTIVE: This study reports the results of a steerable delivery system under the guidance of transesophageal echocardiography (TEE) for the treatment of transjugular closure of secundum atrial septal defects (ASD). METHODS: From July 2015 to May 2016, 33 patients underwent transjugular closure of a secundum ASD under general anesthesia with TEE guidance. The right internal jugular vein was punctured and a FuStar™ steerable sheath was implanted into the right atrium and aligned vertically with the septum, and a closure device was deployed to close the defect. RESULTS: Thirty-two patients with an ASD were successfully occluded; one patient required ASD closure on cardiopulmonary bypass. Procedure time ranged from 5 to 15 (8.2 ± 3.8) min. The length of stay was three to five days after the operation. The follow-up time ranged from 1.1 to 11.0 (5.5 ± 1.5) months. There was no valve regurgitation, no malignant arrhythmias, no device dislocation, or other serious complications. CONCLUSIONS: A steerable delivery system under the guidance of TEE is a safe, effective, and cosmetic method for the transjugular closure of secundum ASDs.


Asunto(s)
Cateterismo Cardíaco/métodos , Procedimientos Quirúrgicos Cardíacos/métodos , Defectos del Tabique Interatrial/cirugía , Dispositivo Oclusor Septal , Cirugía Asistida por Computador/métodos , Adolescente , Adulto , Niño , Preescolar , Ecocardiografía Transesofágica , Femenino , Estudios de Seguimiento , Defectos del Tabique Interatrial/diagnóstico , Humanos , Venas Yugulares , Masculino , Estudios Retrospectivos , Adulto Joven
13.
J Biol Chem ; 289(29): 20193-9, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24895131

RESUMEN

Alcohol dehydrogenase 5 (ADH5) is a conserved enzyme for alcohol and aldehyde metabolism in mammals. Despite dynamic expression throughout neurogenesis, its role in neuronal development remains unknown. Here we present the first evidence that ADH5 is a negative regulator of neuronal differentiation. Gene expression analyses identify a constant reduction of ADH5 levels throughout neuronal development. Overexpression of ADH5 reduces both development and adult neuronal differentiation of mouse neurons. This effect depends on the catalytic activity of ADH5 and involves ADH5-mediated denitrosation of histone deacetylase 2 (HDAC2). Our results indicate that ADH5 counteracts neuronal differentiation of human neural stem cells and that this effect can be reversed by pharmacological inhibition of ADH5. Based on these observations, we propose that ADH5 is a novel suppressor of neuronal differentiation and maturation. Inhibition of ADH5 may improve adult neurogenesis in a physiological or pathological setting.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Neuronas/citología , Neuronas/enzimología , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hipocampo/citología , Hipocampo/enzimología , Histona Desacetilasa 2/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/citología , Células-Madre Neurales/enzimología , Neurogénesis/genética , Neurogénesis/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
14.
Free Radic Res ; 58(2): 117-129, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38385781

RESUMEN

Ferroptosis is recognized as a new type of regulated cell death initiated by iron-dependent accumulation of lipid peroxidation. Recent studies have shown that the administration of ascorbic acid (AA) preferentially kills tumor cells by impairing iron metabolism and exerting pro-oxidant effects. Despite mounting evidence indicating the anticancer potential of AA, the underlying molecular mechanisms remain unknown. In this study, we demonstrated that AA decreased cell viability and Ki67 expression, along with its accumulation in the G0/G1 phase in FaDu and SCC-154 cell lines. Furthermore, AA exposure induced morphological changes in mitochondria associated with ferroptosis. AA-induced ferroptosis is accompanied by depletion of glutathione (GSH) and increased levels of ferrous ions (Fe2+), reactive oxygen species (ROS), and malondialdehyde (MDA). However, these ferroptotic effects were ameliorated by deferoxamine and N-acetylcysteine. Network pharmacology results showed that signal transducer and activator of transcription 3 (STAT3) is a key target of AA against oropharyngeal cancer. AA markedly downregulates the relative mRNA expression of STAT3 and glutathione peroxidase 4 (GPX4). Immunoblotting indicated that the protein levels of p-STAT3, STAT3, and GPX4 in FaDu and SCC-154 cells decreased significantly in response to AA treatment. Mechanistically, a chromatin immunoprecipitation assay confirmed that AA exposure reduced STAT3 expression in the GPX4 promoter region. Additionally, AA-induced inhibition of cell growth and ferroptosis was suppressed by STAT3 and GPX4 overexpression, respectively. In summary, AA inhibited oropharyngeal cancer cell growth in vitro by regulating STAT3/GPX4-mediated ferroptosis, which may provide a novel theoretical basis for the clinical treatment of oropharyngeal cancer with AA.


Ascorbic acid acts as an anticancer agent by inducing ferroptosis, reducing the viability of SCC-154 and FaDu cells.Ascorbic acid-mediated ferroptosis acts through STAT3/GPX4 pathway.The induction of ferroptosis has a significant potential for cancer therapy.


Asunto(s)
Ferroptosis , Neoplasias Orofaríngeas , Humanos , Ácido Ascórbico/farmacología , Factor de Transcripción STAT3/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hierro/metabolismo
15.
Pharmacol Ther ; 256: 108610, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38367868

RESUMEN

Accumulating evidence indicates that epigenetic events undergo deregulation in various cancer types, playing crucial roles in tumor development. Among the epigenetic factors involved in the epigenetic remodeling of chromatin, the chromodomain helicase DNA-binding protein (CHD) family frequently exhibits gain- or loss-of-function mutations in distinct cancer types. Therefore, targeting CHD remodelers holds the potential for antitumor treatment. In this review, we discuss epigenetic regulations of cancer development. We emphasize proteins in the CHD family, delving deeply into the intricate mechanisms governing their functions. Additionally, we provide an overview of current therapeutic strategies targeting CHD family members in preclinical trials. We further discuss the promising approaches that have demonstrated early signs of success in cancer treatment.


Asunto(s)
Cromatina , Neoplasias , Humanos , ADN Helicasas/genética , ADN Helicasas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Mutación , Proteínas de Unión al ADN/metabolismo , Ensamble y Desensamble de Cromatina , Epigénesis Genética
16.
BMJ Open ; 14(3): e071821, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38485471

RESUMEN

OBJECTIVES: To develop an interpretable deep learning model of lupus nephritis (LN) relapse prediction based on dynamic multivariable time-series data. DESIGN: A single-centre, retrospective cohort study in China. SETTING: A Chinese central tertiary hospital. PARTICIPANTS: The cohort study consisted of 1694 LN patients who had been registered in the Nanjing Glomerulonephritis Registry at the National Clinical Research Center of Kidney Diseases, Jinling Hospital from January 1985 to December 2010. METHODS: We developed a deep learning algorithm to predict LN relapse that consists of 59 features, including demographic, clinical, immunological, pathological and therapeutic characteristics that were collected for baseline analysis. A total of 32 227 data points were collected by the sliding window method and randomly divided into training (80%), validation (10%) and testing sets (10%). We developed a deep learning algorithm-based interpretable multivariable long short-term memory model for LN relapse risk prediction considering censored time-series data based on a cohort of 1694 LN patients. A mixture attention mechanism was deployed to capture variable interactions at different time points for estimating the temporal importance of the variables. Model performance was assessed according to C-index (concordance index). RESULTS: The median follow-up time since remission was 4.1 (IQR, 1.7-6.7) years. The interpretable deep learning model based on dynamic multivariable time-series data achieved the best performance, with a C-index of 0.897, among models using only variables at the point of remission or time-variant variables. The importance of urinary protein, serum albumin and serum C3 showed time dependency in the model, that is, their contributions to the risk prediction increased over time. CONCLUSIONS: Deep learning algorithms can effectively learn through time-series data to develop a predictive model for LN relapse. The model provides accurate predictions of LN relapse for different renal disease stages, which could be used in clinical practice to guide physicians on the management of LN patients.


Asunto(s)
Aprendizaje Profundo , Nefritis Lúpica , Humanos , Nefritis Lúpica/diagnóstico , Nefritis Lúpica/tratamiento farmacológico , Estudios de Cohortes , Estudios Retrospectivos , Recurrencia
17.
JTCVS Tech ; 17: 133-137, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36820343

RESUMEN

Objectives: To assess the mid-term safety and efficacy of transthoracic perimembranous ventricular septal defect (Pm-VSD) closure using a new biodegradable device. Implantation entailed right subaxillary minithoracotomy under transesophageal echocardiography guidance. Methods: Between October 2019 and January 2020, 13 patients (males, 5; mean age, 3.6 ± 2.5 years) with Pm-VSDs underwent transthoracic device closures at Zhengzhou University Central China Fuwai Hospital as described previously. Delivery pathways were established by manipulating a hollow probe from right atrium through tricuspid valve to right ventricle and then through VSDs to left ventricle, whereupon installation took place. Results: All occluder implantations were successfully executed. Mean defect size was 4.1 ± 1.0 mm, and mean device waist size was 5.2 ± 1.1 mm. One patient (7.7%) with 1.5-mm residual shunt showed complete closure at discharge. There was 1 instance of postoperative incomplete right bundle branch block, which converted to complete right bundle branch block at month 1. During patient follow-up (mean, 24.6 ± 0.8 months), no device dislocations, new residual shunts, new valvular regurgitation, or detectable atrioventricular block ensued. Conclusions: Closure of Pm-VSDs using a novel, fully biodegradable occluder in the manner described has proven safe and effective at mid-term follow-up. Long-term safety and efficacy of this device must be further corroborated in a large patient cohort going forward.

18.
Cancers (Basel) ; 14(24)2022 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-36551720

RESUMEN

Accurate detection of HER2 expression through immunohistochemistry (IHC) is of great clinical significance in the treatment of breast cancer. However, manual interpretation of HER2 is challenging, due to the interobserver variability among pathologists. We sought to explore a deep learning method to predict HER2 expression level and gene status based on a Whole Slide Image (WSI) of the HER2 IHC section. When applied to 228 invasive breast carcinoma of no special type (IBC-NST) DAB-stained slides, our GrayMap+ convolutional neural network (CNN) model accurately classified HER2 IHC level with mean accuracy 0.952 ± 0.029 and predicted HER2 FISH status with mean accuracy 0.921 ± 0.029. Our result also demonstrated strong consistency in HER2 expression score between our system and experienced pathologists (intraclass correlation coefficient (ICC) = 0.903, Cohen's κ = 0.875). The discordant cases were found to be largely caused by high intra-tumor staining heterogeneity in the HER2 IHC group and low copy number in the HER2 FISH group.

19.
Clin Transl Med ; 12(5): e852, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35538890

RESUMEN

BACKGROUND: Glutaminolysis is a critical metabolic process that promotes cancer cell proliferation, including hepatocellular carcinoma (HCC). Delineating the molecular control of glutaminolysis could identify novel targets to ameliorate this oncogenic metabolic pathway. Here, we evaluated the role of general control of amino acid synthesis 5 like 1 (GCN5L1), a regulator of mitochondrial protein acetylation, in modulating the acetylation and activity of glutaminase to regulate HCC development. METHODS: Cell proliferation was determined by MTT, 2D and soft agar clone formation assays and orthotopic tumour assays in nude mice. GLS1/2 acetylation and activities were measured in cells and tumours to analyse the correlation with GCN5L1 expression and mTORC1 activation. RESULTS: Hepatic GCN5L1 ablation in mice markedly increased diethylnitrosamine (DEN)-induced HCC, and conversely, the transduction of mitochondrial-restricted GCN5L1 protected wild-type mice against HCC progression in response to DEN and carbon tetrachloride (CCl4 ) exposure. GCN5L1-depleted HepG2 hepatocytes enhanced tumour growth in athymic nude mice. Mechanistically, GCN5L1 depletion promoted cell proliferation through mTORC1 activation. Interestingly, liver-enriched glutaminase 2 (GLS2) appears to play a greater role than ubiquitous and canonical tumour-enriched glutaminase 1 (GLS1) in promoting murine HCC. Concurrently, GCN5L1 promotes acetylation and inactivation of both isoforms and increases enzyme oligomerisation. In human HCC tumours compared to adjacent tissue, there were variable levels of mTORC1 activation, GCN5L1 levels and glutaminase activity. Interestingly, the levels of GCN5L1 inversely correlated with mTORC1 activity and glutaminase activity in these tumours. CONCLUSIONS: Our study identified that glutaminase activity, rather than GLS1 or GLS2 expression, is the key factor in HCC development that activates mTORC1 and promotes HCC. In the Kaplan-Meier analysis of liver cancer, we found that HCC patients with high GCN5L1 expression survived longer than those with low GCN5L1 expression. Collectively, GCN5L1 functions as a tumour regulator by modulating glutaminase acetylation and activity in the development of HCC.


Asunto(s)
Carcinoma Hepatocelular , Glutaminasa , Neoplasias Hepáticas , Proteínas Mitocondriales , Proteínas del Tejido Nervioso , Acetilación , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Glutaminasa/genética , Glutaminasa/metabolismo , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Desnudos , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo
20.
Stem Cell Res ; 64: 102905, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36070637

RESUMEN

Genetic studies show that BLOC1S1 modulates mitochondrial and endosome-lysosome function (Wu et al., 2021a). Furthermore, Bloc1s1 mutations are linked to leukodystrophy (Bertoli-Avella et al., 2021). The Vanderver laboratory identified additional individuals with leukodystrophy that harbored either complex heterozygous (Bloc1s1 c.206A > C and c.359G > A), or homozygous (Bloc1s1 c.185 T > C) point mutations. We generated induced pluripotential stem cell (iPSC) lines from these subjects, from parents of the complex heterozygous mutations patient, and from CRISPR isogenic (c.206A > C and c.359G > A) corrected iPSC-line. These complex heterozygous, homozygous, and isogenic-corrected Bloc1s1 lines were phenotypically normal and were capable of differentiation towards the three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Homocigoto , Células Madre Pluripotentes Inducidas/metabolismo , Heterocigoto , Mutación/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteínas del Tejido Nervioso/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA