Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 127, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38782919

RESUMEN

DEAD-box helicase 17 (DDX17) is a typical member of the DEAD-box family with transcriptional cofactor activity. Although DDX17 is abundantly expressed in the myocardium, its role in heart is not fully understood. We generated cardiomyocyte-specific Ddx17-knockout mice (Ddx17-cKO), cardiomyocyte-specific Ddx17 transgenic mice (Ddx17-Tg), and various models of cardiomyocyte injury and heart failure (HF). DDX17 is downregulated in the myocardium of mouse models of heart failure and cardiomyocyte injury. Cardiomyocyte-specific knockout of Ddx17 promotes autophagic flux blockage and cardiomyocyte apoptosis, leading to progressive cardiac dysfunction, maladaptive remodeling and progression to heart failure. Restoration of DDX17 expression in cardiomyocytes protects cardiac function under pathological conditions. Further studies showed that DDX17 can bind to the transcriptional repressor B-cell lymphoma 6 (BCL6) and inhibit the expression of dynamin-related protein 1 (DRP1). When DDX17 expression is reduced, transcriptional repression of BCL6 is attenuated, leading to increased DRP1 expression and mitochondrial fission, which in turn leads to impaired mitochondrial homeostasis and heart failure. We also investigated the correlation of DDX17 expression with cardiac function and DRP1 expression in myocardial biopsy samples from patients with heart failure. These findings suggest that DDX17 protects cardiac function by promoting mitochondrial homeostasis through the BCL6-DRP1 pathway in heart failure.


Asunto(s)
ARN Helicasas DEAD-box , Insuficiencia Cardíaca , Miocitos Cardíacos , Animales , Humanos , Ratones , Apoptosis/genética , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/metabolismo , Homeostasis/genética , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Dinámicas Mitocondriales/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo
2.
Planta ; 259(3): 57, 2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38307982

RESUMEN

MAIN CONCLUSION: This review summarizes the physiological, biochemical, and molecular regulatory network changes in plants in response to high temperature. With the continuous rise in temperature, high temperature has become an important issue limiting global plant growth and development, affecting the phenotype and physiological and biochemical processes of plants and seriously restricting crop yield and tree growth speed. As sessile organisms, plants inevitably encounter high temperatures and improve their heat tolerance by activating molecular networks related to heat stress, such as signal transduction, synthesis of metabolites, and gene expression. Heat tolerance is a polygenic trait regulated by a variety of genes, transcription factors, proteins, and metabolites. Therefore, this review summarizes the changes in physiological, biochemical and molecular regulatory networks in plants under high-temperature conditions to lay a foundation for an in-depth understanding of the mechanisms involved in plant heat tolerance responses.


Asunto(s)
Respuesta al Choque Térmico , Plantas , Temperatura , Plantas/genética , Plantas/metabolismo , Respuesta al Choque Térmico/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fenotipo , Estrés Fisiológico , Regulación de la Expresión Génica de las Plantas , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
3.
Diagnostics (Basel) ; 13(17)2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37685269

RESUMEN

BACKGROUND: Age-related macular degeneration (AMD) is a progressive ocular ailment causing age-associated vision deterioration, characterized by dysregulated immune cell activity. Notably, follicular helper T (Tfh) cells have emerged as pivotal contributors to AMD pathogenesis. Nonetheless, investigations into Tfh-associated gene biomarkers for this disorder remain limited. METHODS: Utilizing gene expression data pertinent to AMD procured from the Gene Expression Omnibus (GEO) repository, we employed the "DESeq2" R software package to standardize and preprocess expression levels. Concurrently, CIBERSORT analysis was utilized to compute the infiltration proportions of 22 distinct immune cell types. Subsequent to weighted gene correlation network analysis (WGCNA), coupled with differential expression scrutiny, we pinpointed genes intricately linked with Tfh cells. These potential genes underwent further screening using the MCODE function within Cytoscape software. Ultimately, a judicious selection of pivotal genes from these identified clusters was executed through the LASSO algorithm. Subsequently, a diagnostic nomogram was devised based on these selected genes. RESULTS: Evident Tfh cell disparities between AMD and control cohorts were observed. Our amalgamated analysis, amalgamating differential expression data with co-expression patterns, unveiled six genes closely associated with Tfh cells in AMD. Subsequent employment of the LASSO algo-rithm facilitated identification of the most pertinent genes conducive to predictive modeling. From these, GABRB3, MFF, and PROX1 were elected as prospective diagnostic biomarkers for AMD. CONCLUSIONS: This investigation discerned three novel biomarker genes, linked to inflammatory mechanisms and pivotal in diagnosing AMD. Further exploration of these genes holds potential to foster novel therapeutic modalities and augment comprehension of AMD's disease trajectory.

4.
Water Res ; 238: 120016, 2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37146397

RESUMEN

Anammox bacteria rely heavily on iron and have many iron storage sites. However, the biological significance of these iron storage sites has not been clearly defined. In this study, we explored the properties and location of iron storage sites to better understand their cellular function. To do this, the Candidatus Kuenenia stuttgartiensis iron storage protein, bacterioferritin (K.S Bfr), was successfully expressed and purified. In vitro, correctly assembled globulins were observed by transmission electron microscopy. The self-assembled K.S Bfr has active redox and can bind Fe2+ and mineralize it in the protein cavity. In vivo, engineered bacteria with K.S Bfr showed good adaptability to Fe2+, with a survival rate of 78.9% when exposed to 5 mM Fe2+, compared with only 66.0% for wild-type bacteria lacking K.S Bfr. A potential iron regulatory strategy similar to that of Anammox was identified in transcriptomic analysis of engineered bacteria. This system may be controlled by the iron uptake regulator Furto transport Fe2+ via FeoB and store excess Fe2+ in K.S Bfr to maintain cellular homeostasis. K.S Bfr has superior iron storage capacity both intracellularly and in vitro. The discovery of K.S Bfr reveals the storage location of iron-rich nanoparticles, increases our understanding of the adaptability of iron-dependent bacteria to Fe2+, and suggests possible iron regulation strategies in Anammox bacteria.


Asunto(s)
Ferritinas , Hierro , Hierro/metabolismo , Ferritinas/química , Ferritinas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacterias/metabolismo , Oxidación-Reducción , Homeostasis
5.
J Hazard Mater ; 440: 129787, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36007364

RESUMEN

Hydroxylamine (NH2OH) is a potentially toxic pollutant when it is present in water, as it can damage both bacteria and the human body. It is still difficult to eliminate the toxic NH2OH in water. Here, we showed that the model bacterium (Escherichia coli) with nanocompartments encapsulated with hydroxylamine oxidase (HAO) can remove NH2OH from water. In addition, the removal efficiency of NH2OH by genetically modified bacteria (with HAO-nanocompartments) was 3.87 mg N L-1 h-1, and that of wild-type bacteria (without HAO-nanocompartments) was only 1.86 mg N L-1 h-1. Label-free quantitative proteomics indicated that the nanocompartments containing HAO enhanced bacterial activity by inducing the up-regulation of proteins involved in stress and stimulus responses, and decreased their intracellular NH2OH concentration. Moreover, the synthesis of proteins involved in energy metabolism, gene expression, and other processes in bacterial was enhanced under hydroxylamine stress, and these changes increased the resistance of bacterial to NH2OH. This work can aid our understanding of the toxic effects of NH2OH on bacteria as well as the development of new approaches to eliminate NH2OH in water.


Asunto(s)
Hidroxilamina , Oxidorreductasas , Contaminantes Químicos del Agua , Bacterias/metabolismo , Hidroxilamina/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Proteómica , Contaminantes Químicos del Agua/metabolismo
6.
J Hazard Mater ; 435: 128954, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35462189

RESUMEN

Zn2+ is largely discharged from many industries and poses a severe threat to the environment, making its remediation crucial. Encapsulins, proteinaceous nano-compartments, may protect cells against environmental stresses by sequestering toxic substances. To determine whether hemerythrin-containing encapsulins (cEnc) from anammox bacteria Ca. Brocadia fulgida can help cells deal with toxic substances such as Zn2+, we transferred cEnc into E.coli by molecular biology technologies for massive expression and then cultured them in media with increasing Zn2+ levels. The engineered bacteria (with cEnc) grew better and entered the apoptosis phase later, while wild bacteria showed poor survival. Furthermore, tandem mass tag-based quantitative proteomic analysis was used to reveal the underlying regulatory mechanism by which the genetically-engineered bacteria (with cEnc) adapted to Zn2+ stress. When Zn2+ was sequestered in cEnc as a transition, the engineered bacteria presented a complex network of regulatory systems against Zn2+-induced cytotoxicity, including functions related to ribosomes, sulfur metabolism, flagellar assembly, DNA repair, protein synthesis, and Zn2+ efflux. Our findings offer an effective and promising stress control strategy to enhance the Zn2+ tolerance of bacteria for Zn2+ remediation and provide a new application for encapsulins.


Asunto(s)
Bacterias , Proteómica , Bacterias/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Oxidación-Reducción , Zinc/metabolismo , Zinc/toxicidad
7.
Oxid Med Cell Longev ; 2022: 5184135, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186188

RESUMEN

OBJECTIVE: To investigate the protective effects and regulatory mechanism of miR-488-3p on doxorubicin-induced cardiotoxicity. METHODS: The C57BL/6 mice and primary cardiomyocytes were used to construct doxorubicin-induced cardiomyocyte injury models in vivo and in vitro. The levels of miR-488-3p and its downstream target genes were analyzed by quantitative real-time PCR. Mouse cardiac function, cell survival, cellular injury-related proteins, and the apoptosis level of cardiomyocytes were analyzed by echocardiography, MTT analysis, Western blotting, and DNA laddering separately. RESULTS: Cardiomyocyte injury caused by a variety of stimuli can lead to the reduction of miR-488-3p level, especially when stimulated with doxorubicin. Doxorubicin led to significant decrease in cardiac function, cell autophagic flux blockage, and apoptosis in vivo and in vitro. The expression of miR-488-3p's target gene, CyclinG1, increased remarkably in the doxorubicin-treated neonatal mouse cardiomyocytes. Overexpression of miR-488-3p inhibited CyclinG1 expression, increased cardiomyocyte viability, and attenuated doxorubicin-induced cardiomyocyte autophagic flux blockage and apoptosis. CONCLUSIONS: miR-488-3p is one of the important protective miRNAs in doxorubicin-induced cardiotoxicity by inhibiting the expression of CyclinG1, which provides insight into the possible clinical application of miR-488-3p/CyclinG1 as therapeutic targets in doxorubicin-induced cardiovascular diseases.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiotoxicidad/etiología , Ciclina G1/antagonistas & inhibidores , Doxorrubicina/efectos adversos , MicroARNs/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Animales , Humanos , Masculino , Ratones , Ratas
8.
J Immunol Res ; 2021: 1815098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34307691

RESUMEN

Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.


Asunto(s)
Adiponectina/metabolismo , Daño por Reperfusión Miocárdica/inmunología , Miocitos Cardíacos/patología , Animales , Animales Recién Nacidos , Apoptosis/inmunología , Hipoxia de la Célula/inmunología , Supervivencia Celular , Células Cultivadas , Medios de Cultivo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/inmunología , Necroptosis/inmunología , Estrés Oxidativo/inmunología , Embarazo , Cultivo Primario de Células , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
Biomed Res Int ; 2021: 8880179, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33532500

RESUMEN

Regulated necrosis (necroptosis) is crucially involved in cardiac ischaemia-reperfusion injury (MIRI). The aim of our study is to investigate whether shock wave therapy (SWT) is capable of exerting protective effects by inhibiting necroptosis during myocardial ischaemia-reperfusion (I/R) injury and the possible role of autophagy in this process. We established a hypoxia/reoxygenation (H/R) model in vitro using HL-1 cells to simulate MIRI. MTS assays and LDH cytotoxicity assay were performed to measure cell viability and cell damage. Annexin V/PI staining was used to determine apoptosis and necrosis. Western blotting was performed to assess the changes in cell signaling pathways associated with autophagy, necroptosis, and apoptosis. Reactive oxygen species (ROS) production was detected using DHE staining. Autophagosome generation and degradation (autophagic flux) were analysed using GFP and RFP tandemly tagged LC3 (tfLC3). HL-1 cells were then transfected with p62/SQSTM1 siRNA in order to analyse its role in cardioprotection. Our results revealed that SWT increased cell viability in the H/R model and decreased receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 expression. ROS production was also inhibited by SWT. Moreover, SWT decreased Beclin1 expression and the ratio of LC3-II/LC3-I following H/R. Simultaneously, in the tfLC3 assay, the SWT provoked a decrease in the cumulative autophagosome abundance. siRNA-mediated knockdown of p62 attenuated H/R-induced necroptosis, and SWT did not exert additive effects. Taken together, SWT ameliorated H/R injury by inhibiting necroptosis. SWT also relieved the blockade of autophagic flux in response to H/R injury. The restoration of autophagic flux by SWT might contribute to its cardioprotective effect on necroptosis following H/R injury.


Asunto(s)
Autofagia/efectos de la radiación , Hipoxia de la Célula/efectos de la radiación , Tratamiento con Ondas de Choque Extracorpóreas , Miocitos Cardíacos , Necroptosis/efectos de la radiación , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Corazón/efectos de la radiación , Ratones , Modelos Biológicos , Daño por Reperfusión Miocárdica , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de la radiación
10.
Acta Pharmacol Sin ; 42(10): 1653-1664, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33441995

RESUMEN

Rheumatoid arthritis (RA) is characterized by joint leukocyte infiltration, synovial inflammation and bone damage result from osteoclastogenesis. Bruton's tyrosine kinase (BTK) is a key regulator of B cell receptor (BCR) and Fc gamma receptor (FcγR) signaling involved in the pathobiology of RA and other autoimmune disorders. SOMCL-17-016 is a potent and selective tricyclic BTK inhibitor, structurally distinct from other known BTK inhibitors. In present study we investigated the therapeutic efficacy of SOMCL-17-016 in a mouse collagen-induced arthritis (CIA) model and underlying mechanisms. CIA mice were administered SOMCL-17-016 (6.25, 12.5, 25 mg·kg-1·d-1, ig), or ibrutinib (25 mg·kg-1·d-1, ig) or acalabrutinib (25 mg·kg-1·d-1, ig) for 15 days. We showed that oral administration of SOMCL-17-016 dose-dependently ameliorated arthritis severity and bone damage in CIA mice; it displayed a higher in vivo efficacy than ibrutinib and acalabrutinib at the corresponding dosage. We found that SOMCL-17-016 administration dose-dependently inhibited anti-IgM-induced proliferation and activation of B cells from CIA mice, and significantly decreased anti-IgM/anti-CD40-stimulated RANKL expression in memory B cells from RA patients. In RANKL/M-CSF-stimulated RAW264.7 cells, SOMCL-17-016 prevented osteoclast differentiation and abolished RANK-BTK-PLCγ2-NFATc1 signaling. In summary, this study demonstrates that SOMCL-17-016 presents distinguished therapeutic effects in the CIA model. SOMCL-17-016 exerts a dual inhibition of B cell function and osteoclastogenesis, suggesting that it to be a promising drug candidate for RA treatment.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Células B de Memoria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Autoanticuerpos/metabolismo , Inflamación/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos DBA , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Pirimidinas/uso terapéutico , Alcaloides de Pirrolicidina/uso terapéutico , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Biomed Res Int ; 2020: 5107193, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32190669

RESUMEN

The usage of doxorubicin is hampered by its life-threatening cardiotoxicity in clinical practice. Dexrazoxane is the only cardioprotective medicine approved by the FDA for preventing doxorubicin-induced cardiac toxicity. Nevertheless, the mechanism of dexrazoxane is incompletely understood. The aim of our study is to investigate the possible molecular mechanism of dexrazoxane against doxorubicin-induced cardiotoxicity. We established a doxorubicin-induced mouse and cardiomyocyte injury model. Male C57BL/6J mice were randomly distributed into a control group (Con), a doxorubicin treatment group (DOX), a doxorubicin plus dexrazoxane treatment group (DOX+DEX), and a dexrazoxane treatment group (DEX). Echocardiography and histology analyses were performed to evaluate heart function and structure. DNA laddering, qRT-PCR, and Western blot were performed on DOX-treated cardiomyocytes with/without DEX treatment in vitro. Cardiomyocytes were then transfected with miR-17-5p mimics or inhibitors in order to analyze its downstream target. Our results demonstrated that dexrazoxane has a potent effect on preventing cardiac injury induced by doxorubicin in vivo and in vitro by reducing cardiomyocyte apoptosis. MicroRNA plays an important role in cardiovascular diseases. Our data revealed that dexrazoxane could upregulate the expression of miR-17-5p, which plays a cytoprotective role in response to hypoxia by regulating cell apoptosis. Furthermore, the miRNA and protein analysis revealed that miR-17-5p significantly attenuated phosphatase and tensin homolog (PTEN) expression in cardiomyocytes exposed to doxorubicin. Taken together, dexrazoxane might exert a cardioprotective effect against doxorubicin-induced cardiomyocyte apoptosis by regulating the expression of miR-17-5p/PTEN cascade.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexrazoxano/farmacología , Doxorrubicina/efectos adversos , MicroARNs/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/patología , Supervivencia Celular/efectos de los fármacos , Dexrazoxano/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfohidrolasa PTEN/metabolismo , Regulación hacia Arriba/efectos de los fármacos
12.
Mol Med Rep ; 21(2): 631-640, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31974607

RESUMEN

Cardiac shock wave therapy (SWT) has been described as a novel therapeutic strategy that is able to alleviate myocardial ischemic injury. microRNA (miRNA/miR)­210 plays a cytoprotective role in cardiomyocytes in response to hypoxia by regulating cell apoptosis. The aim of the present study was to investigate whether cardiac SWT could protect cardiomyocytes from hypoxia­induced injury by regulating miR­210 expression. The murine adult cardiomyocyte cell line HL­1 was incubated for 5 h in hypoxic conditions, followed by reoxygenation for 12 h and treatment with SWT immediately following hypoxia in the present study. The cell viability was determined using an MTS assay. Western blot analyses were performed in order to detect cell signaling changes. Reactive oxygen species production was detected using dihydroethidium staining, and malondialdehyde levels were measured using the thiobarbituric acid method. miRNA and mRNA expression levels were confirmed via reverse transcription­quantitative PCR. Apoptosis was evaluated by means of flow cytometry. HL­1 cells were then transfected with miR­210 mimics or inhibitors in order to alter miR­210 expression levels, and the effects on HL­1 cells were determined. Hypoxia led to elevated oxidative stress, enhanced cell apoptosis and upregulated miR­210 expression levels in HL­1 cells, while SWT could alleviate hypoxia­induced cell injury and further promote miR­210 expression. miR­210 overexpression decreased apoptosis and oxidative stress during hypoxic stress in HL­1 cells, whereas inhibition of miR­210 increased cell apoptosis and promoted oxidative stress. Furthermore, miR­210 inhibition could reverse the effects of SWT on HL­1 cells. Finally, the mRNA analysis revealed that SWT significantly attenuated apoptosis­inducing factor mitochondrion­associated 3 and caspase 8 associated protein 2 mRNA expression levels in cardiomyocytes exposed to hypoxia, which were two targets of miR­210. SWT could exert cardioprotective effects against hypoxia­induced cardiac injury by modulating miR­210.


Asunto(s)
Citoprotección , Tratamiento con Ondas de Choque Extracorpóreas , MicroARNs/metabolismo , Miocitos Cardíacos/patología , Animales , Apoptosis/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Hipoxia de la Célula/genética , Línea Celular , Citoprotección/genética , Ratones , MicroARNs/genética , Estrés Oxidativo/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Biochem Biophys Res Commun ; 523(1): 140-146, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31837803

RESUMEN

Doxorubicin, as a first line chemotherapeutic agent, its usage is limited owing to cardiotoxicity. Necroptosis is a new form of programmed cell death, and recent investigations indicated that necroptosis is vitally involved in serious cardiac pathological conditions. Dexrazoxane is the only cardiac protective drug approved by FDA for anthracycline. We aimed to explore whether and how dexrazoxane regulates doxorubicin-induced cardiomyocyte necroptosis. First, doxorubicin could cause heart failure and reduce cardiomyocyte viability by promoting cell apoptosis and necroptosis in vivo and in vitro. Second, necroptosis plays an important role in doxorubicin induced cardiomyocyte injury, which could be inhibited by Nec-1. Third, dexrazoxane increased cell viability and protect heart function by decreasing both cardiomyocyte apoptosis and necroptosis after doxorubicin treatment. Forth, dexrazoxane attenuated doxorubicin-induced inflammation and necroptosis by the inhibition of p38MAPK/NF-κB pathways. These results indicated that dexrazoxane ameliorates cardiotoxicity and protects heart function by attenuating both apoptosis and necroptosis in doxorubicin induced cardiomyocyte injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexrazoxano/farmacología , Doxorrubicina/efectos adversos , Miocitos Cardíacos/efectos de los fármacos , Necroptosis/efectos de los fármacos , Animales , Células Cultivadas , Dexrazoxano/administración & dosificación , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
14.
Oxid Med Cell Longev ; 2019: 8768164, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31612078

RESUMEN

Chronic hypertension, valvular heart disease, and heart infarction cause cardiac remodeling and potentially lead to a series of pathological and structural changes in the left ventricular myocardium and a progressive decrease in heart function. Angiotensin II (AngII) plays a key role in the onset and development of cardiac remodeling. Many microRNAs (miRNAs), including miR-154-5p, may be involved in the development of cardiac remolding, but the underlying molecular mechanisms remain unclear. We aimed to characterize the function of miR-154-5p and reveal its mechanisms in cardiac remodeling induced by AngII. First, angiotensin II led to concurrent increases in miR-154-5p expression and cardiac remodeling in adult C57BL/6J mice. Second, overexpression of miR-154-5p to a level similar to that induced by AngII was sufficient to trigger cardiomyocyte hypertrophy and apoptosis, which is associated with profound activation of oxidative stress and inflammation. Treatment with a miR-154-5p inhibitor noticeably reversed these changes. Third, miR-154-5p directly inhibited arylsulfatase B (Arsb) expression by interacting with its 3'-UTR and promoted cardiomyocyte hypertrophy and apoptosis. Lastly, the angiotensin type 1 receptor blocker telmisartan attenuated AngII-induced cardiac hypertrophy, apoptosis, and fibrosis by blocking the increase in miR-154-5p expression. Moreover, upon miR-154-5p overexpression in isolated cardiomyocytes, the protective effect of telmisartan was partially abolished. Based on these results, increased cardiac miR-154-5p expression is both necessary and sufficient for AngII-induced cardiomyocyte hypertrophy and apoptosis, suggesting that the upregulation of miR-154-5p may be a crucial pathological factor and a potential therapeutic target for cardiac remodeling.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones
15.
Cell Physiol Biochem ; 42(2): 673-684, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28618416

RESUMEN

BACKGROUND: Autophagy plays an important role in cardiovascular disease. Controversy still exists regarding the effect of autophagy on ischemic/hypoxic myocardium. Cardiac shock wave therapy (CSWT) is an effective alternative treatment for refractory ischemic heart disease. Whether CSWT can regulate cardiomyocyte autophagy under hypoxic conditions is not clear. We established a myocardial hypoxia model using the H9c2 cell line and performed shock waves (SWs) treatment to evaluate the effect of SW on autophagy. METHODS: The H9c2 cells were incubated under hypoxic conditions, and SW treatment was then performed at energies of 0.02, 0.05, or 0.10 mJ/mm2. The cell viability and intracellular ATP level were examined. Western blot analysis was used to assess the expression of LC3B, AMPK, mTOR, Beclin-1, Sirt1, and HIF-1α. Autophagic vacuoles were visualized by monodansylcadaverine staining. RESULTS: After the 24-hour hypoxic period, cardiomyocyte viability and ATP levels were decreased and autophagy was significantly increased in H9c2 cells. SW treatment with an energy of 0.05 mJ/mm2 significantly increased the cellular viability, ATP level, LC3B-II/I, and number of autophagic vacuoles. In addition, phosphorylated AMPK and Sirt1 were increased and phosphorylated mTOR and HIF-1α were decreased after SW treatment. CONCLUSION: SW treatment can potentially promote cardiomyocyte autophagy during hypoxia and protect cardiomyocyte function by regulating the AMPK/mTOR pathway.


Asunto(s)
Autofagia/efectos de la radiación , Ondas de Choque de Alta Energía/uso terapéutico , Isquemia Miocárdica/terapia , Miocitos Cardíacos/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Hipoxia de la Célula/genética , Línea Celular , Supervivencia Celular/efectos de la radiación , Modelos Animales de Enfermedad , Humanos , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/patología , Fagosomas/metabolismo , Fagosomas/efectos de la radiación , Fosforilación , Ratas , Transducción de Señal/efectos de la radiación
16.
Oncotarget ; 8(3): 4837-4848, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-27902477

RESUMEN

Doxorubicin (adriamycin), an anthracycline antibiotic, is commonly used to treat many types of solid and hematological malignancies. Unfortunately, clinical usage of doxorubicin is limited due to the associated acute and chronic cardiotoxicity. Previous studies demonstrated that Astragalus polysaccharide (APS), the extracts of Astragalus membranaceus, had strong anti-tumor activities and anti-inflammatory effects. However, whether APS could mitigate chemotherapy-induced cardiotoxicity is unclear thus far. We used a doxorubicin-induced neonatal rat cardiomyocyte injury model and a mouse heart failure model to explore the function of APS. GFP-LC3 adenovirus-mediated autophagic vesicle assays, GFP and RFP tandemly tagged LC3 (tfLC3) assays and Western blot analyses were performed to analyze the cell function and cell signaling changes following APS treatment in cardiomyocytes. First, doxorubicin treatment led to C57BL/6J mouse heart failure and increased cardiomyocyte apoptosis, with a disturbed cell autophagic flux. Second, APS restored autophagy in doxorubicin-treated primary neonatal rat ventricular myocytes and in the doxorubicin-induced heart failure mouse model. Third, APS attenuated doxorubicin-induced heart injury by regulating the AMPK/mTOR pathway. The mTOR inhibitor rapamycin significantly abrogated the protective effect of APS. These results suggest that doxorubicin could induce heart failure by disturbing cardiomyocyte autophagic flux, which may cause excessive cell apoptosis. APS could restore normal autophagic flux, ameliorating doxorubicin-induced cardiotoxicity by regulating the AMPK/mTOR pathway.


Asunto(s)
Astragalus propinquus/química , Cardiotoxicidad/tratamiento farmacológico , Doxorrubicina/toxicidad , Insuficiencia Cardíaca/tratamiento farmacológico , Pruebas de Función Cardíaca/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Polisacáridos/administración & dosificación , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Autofagia , Cardiotoxicidad/metabolismo , Cardiotoxicidad/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Polisacáridos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
17.
Angew Chem Int Ed Engl ; 52(17): 4538-43, 2013 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-23564565

RESUMEN

Inside and outside: Two consecutive postsynthetic modifications, first an elimination reaction in the channels and then bromination at the surface, were realized in a new hybrid metal-organic framework. The dramatic effects of the different groups in the channels and at the surface were studied using gas sorption and the loading/release of solvent and iodine.


Asunto(s)
Complejos de Coordinación/síntesis química , Hidrocarburos Bromados/síntesis química , Zinc/química , Absorción , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/química , Complejos de Coordinación/química , Cristalografía por Rayos X , Hidrocarburos Bromados/química , Imidazoles/síntesis química , Imidazoles/química , Modelos Moleculares , Procesos Fotoquímicos , Estereoisomerismo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA