Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1363278, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601160

RESUMEN

Purpose: A mouse model of irradiation (IR)-induced heart injury was established to investigate the early changes in cardiac function after radiation and the role of cardiac macrophages in this process. Methods: Cardiac function was evaluated by heart-to-tibia ratio, lung-to-heart ratio and echocardiography. Immunofluorescence staining and flow cytometry analysis were used to evaluate the changes of macrophages in the heart. Immune cells from heart tissues were sorted by magnetic beads for single-cell RNA sequencing, and the subsets of macrophages were identified and analyzed. Trajectory analysis was used to explore the differentiation relationship of each macrophage subset. The differentially expressed genes (DEGs) were compared, and the related enriched pathways were identified. Single-cell regulatory network inference and clustering (SCENIC) analysis was performed to identify the potential transcription factors (TFs) which participated in this process. Results: Cardiac function temporarily decreased on Day 7 and returned to normal level on Day 35, accompanied by macrophages decreased and increased respectively. Then, we identified 7 clusters of macrophages by single-cell RNA sequencing and found two kinds of stage specific macrophages: senescence-associated macrophage (Cdkn1ahighC5ar1high) on Day 7 and interferon-associated macrophage (Ccr2highIsg15high) on Day 35. Moreover, we observed cardiac macrophages polarized over these two-time points based on M1/M2 and CCR2/major histocompatibility complex II (MHCII) expression. Finally, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses suggested that macrophages on Day 7 were characterized by an inflammatory senescent phenotype with enhanced chemotaxis and inflammatory factors, while macrophages on Day 35 showed enhanced phagocytosis with reduced inflammation, which was associated with interferon-related pathways. SCENIC analysis showed AP-1 family members were associated with IR-induced macrophages changes. Conclusion: We are the first study to characterize the diversity, features, and evolution of macrophages during the early stages in an IR-induced cardiac injury animal model.


Asunto(s)
Macrófagos , Fagocitosis , Ratones , Animales , Inflamación/metabolismo , Interferones/metabolismo , Análisis de Secuencia de ARN
2.
EBioMedicine ; 95: 104744, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37556943

RESUMEN

BACKGROUND: Macrophage infiltration and polarization are integral to the progression of heart failure and cardiac fibrosis after ischemia/reperfusion (IR). Interleukin 34 (IL-34) is an inflammatory regulator related to a series of autoimmune diseases. Whether IL-34 mediates inflammatory responses and contributes to cardiac remodeling and heart failure post-IR remains unclear. METHODS: IL-34 knock-out mice were used to determine the role of IL-34 on cardiac remodeling after IR surgery. Then, immunofluorescence, flow cytometry assays, and RNA-seq analysis were performed to explore the underlying mechanisms of IL-34-induced macrophage recruitment and polarization, and further heart failure after IR. FINDINGS: By re-analyzing single-cell RNA-seq and single-nucleus RNA-seq data of murine and human ischemic hearts, we showed that IL-34 expression was upregulated after IR. IL-34 knockout mitigated cardiac remodeling, cardiac dysfunction, and fibrosis after IR and vice versa. RNA-seq analysis revealed that IL-34 deletion correlated negatively with immune responses and chemotaxis after IR injury. Consistently, immunofluorescence and flow cytometry assays demonstrated that IL-34 deletion attenuated macrophage recruitment and CCR2+ macrophage polarization. Mechanistically, IL-34 deficiency repressed both the canonical and noncanonical NF-κB signaling pathway, leading to marked reduction of P-IKKß and P-IκBα kinase levels; downregulation of NF-κB p65, RelB, and p52 expression, which drove the decline in chemokine CCL2 expression. Finally, IL-34 and CCL2 levels were increased in the serum of acute coronary syndrome patients, with a positive correlation between circulating IL-34 and CCL2 levels in clinical patients. INTERPRETATION: In conclusion, IL-34 sustains NF-κB pathway activation to elicit increased CCL2 expression, which contributes to macrophage recruitment and polarization, and subsequently exacerbates cardiac remodeling and heart failure post-IR. Strategies targeting IL-34-centered immunomodulation may provide new therapeutic approaches to prevent and reverse cardiac remodeling and heart failure in clinical MI patients after percutaneous coronary intervention. FUNDING: This study was supported by the National Nature Science Foundation of China (81670352 and 81970327 to R T, 82000368 to Q F).


Asunto(s)
Insuficiencia Cardíaca , Interleucinas , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , FN-kappa B , Animales , Humanos , Ratones , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Quinasa I-kappa B/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Macrófagos/metabolismo , Ratones Noqueados , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Remodelación Ventricular
3.
Biomolecules ; 13(7)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37509108

RESUMEN

Adverse cardiac remodeling, including cardiac fibrosis, after myocardial infarction (MI) is a major cause of long-term heart failure. 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), an enzyme that regulates glucose metabolism, also plays an important role in various fibrotic and cardiovascular diseases. However, its effects on MI remain unknown. Here, PFKFB3 inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) and a permanent left anterior descending ligation mouse model were used to explore the functional role of PFKFB3 in MI. We showed that PFKFB3 expression increased significantly in the area of cardiac infarction during the early phase after MI, peaking on day 3. 3PO treatment markedly improved cardiac function, accompanied by decreased infarction size and collagen density in the infarct area. Meanwhile, 3PO attenuated cardiac fibrosis after MI by reducing the expression of collagen and fibronectin in murine hearts. Notably, 3PO reduced PFKFB3 expression and inhibited the transforming growth factor-beta 1/mothers against the decapentaplegic homolog 2/3 (TGF-ß1/SMAD2/3) signaling pathway to inhibit cardiac fibrosis after MI. Moreover, PFKFB3 expression in neonatal rat cardiac fibroblasts (NRCFs) increased significantly after MI and under hypoxia, whereas 3PO alleviated the migratory capacity and activation of NRCFs induced by TGF-ß1. In conclusion, 3PO effectively reduced fibrosis and improved adverse cardiac remodeling after MI, suggesting PFKFB3 inhibition as a novel therapeutic strategy to reduce the incidence of chronic heart failure following MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Ratas , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Ventricular , Infarto del Miocardio/metabolismo , Colágeno , Insuficiencia Cardíaca/tratamiento farmacológico , Fibrosis
4.
J Am Heart Assoc ; 11(24): e027228, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36515244

RESUMEN

Background Myocardial infarction (MI) is characterized by the emergence of dead or dying cardiomyocytes and excessive immune cell infiltration after coronary vessel occlusion. However, the complex transcriptional profile, pathways, cellular interactome, and transcriptional regulators of immune subpopulations after MI remain elusive. Methods and Results Here, male C57BL/6 mice were subjected to MI surgery and monitored for 1 day and 7 days, or sham surgery for 7 days, then cardiac CD45-positive immune cells were collected for single-cell RNA sequencing to determine immune heterogeneity. A total of 30 135 CD45+ immune cells were partitioned into macrophages, monocytes, neutrophils, dendritic cells, and T or B cells for further analysis. We showed that macrophages enriched for Olr1 and differentially expressed Gpnmb represented 2 crucial ischemia-associated macrophages with distinct proinflammatory and prophagocytic capabilities. In contrast to the proinflammatory subset of macrophages enriched for Olr1, Gpnmb-positive macrophages exhibited higher phagocytosis and fatty acid oxidation preference, which could be abolished by etomoxir treatment. In addition to macrophages, MI triggered prompt recruitment of neutrophils into murine hearts, which constituted the sequential cell-fate from naïve S100a4-positive, to activated Sell-high, to aging Icam1-high neutrophils. In silico tools predicted that the excessively expanded neutrophils at 1 day were attributed to chemokine C-C motif ligand/chemokine C-X-C motif ligand pathways, whereas CD80/inducible T-cell costimulator (ICOS) signaling was responsible for the immunosuppressive response at day 7 after MI. Finally, the Fos/AP-1 (activator protein 1) regulon was identified as the critical regulator of proinflammatory responses, which was significantly activated in patients with dilated cardiomyopathy and ischemic cardiomyopathy. We showed the enriched Fos/AP-1 target gene loci in genome-wide association study signals for coronary artery diseases and MI. Targeting Fos/AP-1 with the selective inhibitor T5224 blunted leukocyte infiltration and alleviated cardiac dysfunction in the preclinical murine MI model. Conclusions Taken together, this single-cell RNA sequencing data lay the groundwork for the understanding of immune cell heterogeneity and dynamics in murine ischemic hearts. Moreover, Fos/AP-1 inhibition mitigates inflammatory responses and cardiac dysfunction, which might provide potential therapeutic benefits for heart failure intervention after MI.


Asunto(s)
Infarto del Miocardio , Miocardio , Masculino , Ratones , Animales , Miocardio/metabolismo , Estudio de Asociación del Genoma Completo , Ligandos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/uso terapéutico , Ratones Endogámicos C57BL , Infarto del Miocardio/tratamiento farmacológico , Quimiocinas/metabolismo , Modelos Animales de Enfermedad
5.
Front Cardiovasc Med ; 9: 907997, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36247428

RESUMEN

Background and aims: Intestinal flora metabolites are associated with cardiovascular (CV) diseases including heart failure (HF). The carnitine precursor trimethyllysine (TML), which participates in the generation of the atherogenic-related metabolite trimethylamine N-oxide (TMAO), was found to be related to poor prognosis in patients with CV diseases. The aim of the present study was to examine the relationship between TML and stable chronic HF. Methods and results: In total, 956 subjects including 471 stable chronic HF and 485 non-HF patients were enrolled in the present cohort study and subjects with stable HF were followed up for 2.0 ± 1.1 years. Serum levels of TML and TMAO were measured by liquid chromatography mass spectrometry in tandem. TML levels were significantly elevated in patients with HF compared with non-HF patients and were positively correlated with N-terminal pro-brain natriuretic peptide (NTproBNP) levels (r = 0.448, P < 0.001). TML was associated with the presence of HF after adjusting for age, sex, complications, traditional clinical factors, and TMAO (tertile 3 (T3), adjusted odds ratio (OR) 1.93, 95% confidence interval (CI) 1.19-3.13, and P = 0.007). In patients with HF, increased TML levels were associated with a composite endpoint of CV death and HF hospitalization during follow-up (T3, adjusted hazard ratio (HR) 1.93, 95% CI 1.27-2.93, and P = 0.002). Increased TML levels indicated a higher risk of CV death, re-hospitalization, and all-cause mortality. Conclusion: Serum TML levels were associated with the presence and severity of HF in all subjects. High levels of TML can indicate complications and poor prognosis in HF patients.

6.
Front Physiol ; 13: 912739, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846006

RESUMEN

Artificial intelligence is increasingly being used on the clinical electrocardiogram workflows. Few electrocardiograms based on artificial intelligence algorithms have focused on detecting myocardial ischemia using long-term electrocardiogram data. A main reason for this is that interference signals generated from daily activities while wearing the Holter monitor lowered the ability of artificial intelligence to detect myocardial ischemia. In this study, an automatic system combining denoising and segmentation modules was developed to detect the deviation of the ST-segment and J point. We proposed a ECG Bidirectional Transformer network that applied in both denoising and segmentation tasks. The denoising model achieved RMSEde, SNRimp, and PRD values of 0.074, 10.006, and 16.327, respectively. The segmentation model achieved precision, sensitivity (recall), and F1-score of 96.00, 93.06, and 94.51%, respectively. The system's ability to distinguish the depression and elevation of the ST-segment and J point was also verified by cardiologists as well. From our ECG dataset, 103 patients with ST-segment depression and 10 patients with ST-segment elevation were detected with positive predictive values of 80.6 and 60% respectively. Using Holter ECG and transformer-based deep neural networks, we can detect subtle ST-segment changes in noisy ECG signals. This system has the potential to improve the efficacy of daily medicine and to provide a broader population-level screening for asymptomatic myocardial ischemia.

7.
ESC Heart Fail ; 9(4): 2645-2653, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35624536

RESUMEN

AIMS: To explore the associations between serum phenylacetylglutamine (PAGln) and chronic heart failure (HF). METHODS AND RESULTS: Totally 956 subjects were enrolled consecutively from the Department of Cardiovascular Medicine, Ruijin Hospital. Baseline data were obtained from all participants, and 471 stable chronic HF subjects were followed up. Serum PAGln was analysed by liquid chromatography-tandem mass spectrometry. The association between PAGln and basic renal indicators was assessed by simple correlation analysis. Logistic regression analysis was conducted to measure the association between PAGln and HF risk. Event-free survival was determined by Kaplan-Meier curves, and differences in survival were assessed using log-rank tests. Cox proportional hazards analysis was used to assess the prognostic value of PAGln in HF. Serum PAGln levels were increased in patients with chronic HF (3.322 ± 8.220 µM vs. 1.249 ± 1.168 µM, P < 0.001) and were associated with HF after full adjustment [odds ratio (OR), 1.507; 95% confidence interval (CI): 1.213-1.873; P < 0.001]. PAGln levels were correlated with the levels of basic renal indicators. High PAGln levels indicated a high risk of renal dysfunction in HF (OR: 1.853; 95% CI: 1.344-2.556; P < 0.001), and elevated PAGln levels were associated with a high risk of cardiovascular death in patients with chronic HF (HR: 2.049; 95% CI: 1.042-4.029; P = 0.038). CONCLUSIONS: Elevated PAGln levels are an independent risk factor for HF and are associated with a higher risk of cardiovascular death. High PAGln levels could indicate renal dysfunction in HF patients. PAGln can be a valuable indicator of HF.


Asunto(s)
Insuficiencia Cardíaca , Enfermedades Renales , Enfermedad Crónica , Glutamina/análogos & derivados , Humanos , Pronóstico , Factores de Riesgo
8.
Circulation ; 145(11): 829-846, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35235343

RESUMEN

BACKGROUND: Heart failure is a global public health issue that is associated with increasing morbidity and mortality. Previous studies have suggested that mitochondrial dysfunction plays critical roles in the progression of heart failure; however, the underlying mechanisms remain unclear. Because kinases have been reported to modulate mitochondrial function, we investigated the effects of DYRK1B (dual-specificity tyrosine-regulated kinase 1B) on mitochondrial bioenergetics, cardiac hypertrophy, and heart failure. METHODS: We engineered DYRK1B transgenic and knockout mice and used transverse aortic constriction to produce an in vivo model of cardiac hypertrophy. The effects of DYRK1B and its downstream mediators were subsequently elucidated using RNA-sequencing analysis and mitochondrial functional analysis. RESULTS: We found that DYRK1B expression was clearly upregulated in failing human myocardium and in hypertrophic murine hearts, as well. Cardiac-specific DYRK1B overexpression resulted in cardiac dysfunction accompanied by a decline in the left ventricular ejection fraction, fraction shortening, and increased cardiac fibrosis. In striking contrast to DYRK1B overexpression, the deletion of DYRK1B mitigated transverse aortic constriction-induced cardiac hypertrophy and heart failure. Mechanistically, DYRK1B was positively associated with impaired mitochondrial bioenergetics by directly binding with STAT3 to increase its phosphorylation and nuclear accumulation, ultimately contributing toward the downregulation of PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α). Furthermore, the inhibition of DYRK1B or STAT3 activity using specific inhibitors was able to restore cardiac performance by rejuvenating mitochondrial bioenergetics. CONCLUSIONS: Taken together, the findings of this study provide new insights into the previously unrecognized role of DYRK1B in mitochondrial bioenergetics and the progression of cardiac hypertrophy and heart failure. Consequently, these findings may provide new therapeutic options for patients with heart failure.


Asunto(s)
Insuficiencia Cardíaca , Función Ventricular Izquierda , Animales , Cardiomegalia/metabolismo , Metabolismo Energético , Insuficiencia Cardíaca/etiología , Humanos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Volumen Sistólico , Quinasas DyrK
9.
Front Physiol ; 12: 746377, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34621191

RESUMEN

Background and Aims: Acute coronary syndrome (ACS) has become one of the most common causes of disability. It is thus important to identify ACS early in the disease course of patients using novel biomarkers for prompt management. Decorin (DCN) was well-acknowledged for its effect on collagen fibrillogenesis and maintaining tissue integrity. Additionally, DCN could release as secreted proteoglycan under pathological conditions. Hence, we aimed to determine the relationship between serum DCN concentration and ACS. Methods: A total of 388 patients who underwent coronary angiography (CAG) in the cardiovascular center of Ruijin Hospital between June 2016 and December 2017 were enrolled in this study. Blood samples were drawn during CAG surgery to determine the serum DCN level of patients with ACS (n = 210) and control subjects (n = 178) using enzyme-linked immunosorbent assay. Results: We found that the serum DCN levels of ACS patients were elevated compared with those of the control subjects (13.59 ± 0.50 vs. 13.17 ± 0.38, respectively, p < 0.001). Furthermore, the serum DCN level, after being adjusted with other cardiovascular factors, was independently associated with ACS. Moreover, an increased serum DCN level was positively correlated with the number of white blood cells and the level of high-sensitivity C-reactive protein (R = 0.3 and 0.11, respectively). Mechanistically, DCN might have elicited an imbalanced inflammatory response during cardiac ischemia by suppressing the expression of anti-inflammatory genes. Conclusion: Serum DCN is a novel biomarker of ACS and contributes to the increased inflammatory response in ischemic heart disease.

10.
Front Cardiovasc Med ; 8: 722908, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34458345

RESUMEN

Background: Cardiac hypertrophy was accompanied by various cardiovascular diseases (CVDs), and due to the high global incidence and mortality of CVDs, it has become increasingly critical to characterize the pathogenesis of cardiac hypertrophy. We aimed to determine the metabolic roles of fatty acid binding protein 3 (FABP3) on transverse aortic constriction (TAC)-induced cardiac hypertrophy. Methods and Results: Transverse aortic constriction or Ang II treatment markedly upregulated Fabp3 expression. Notably, Fabp3 ablation aggravated TAC-induced cardiac hypertrophy and cardiac dysfunction. Multi-omics analysis revealed that Fabp3-deficient hearts exhibited disrupted metabolic signatures characterized by increased glycolysis, toxic lipid accumulation, and compromised fatty acid oxidation and ATP production under hypertrophic stimuli. Mechanistically, FABP3 mediated metabolic reprogramming by directly interacting with PPARα, which prevented its degradation and synergistically modulated its transcriptional activity on Mlycd and Gck. Finally, treatment with the PPARα agonist, fenofibrate, rescued the pro-hypertrophic effects of Fabp3 deficiency. Conclusions: Collectively, these findings reveal the indispensable roles of the FABP3-PPARα axis on metabolic homeostasis and the development of hypertrophy, which sheds new light on the treatment of hypertrophy.

11.
Front Cardiovasc Med ; 7: 615161, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33365332

RESUMEN

Advances in single-cell RNA sequencing (scRNA-seq) technology have recently shed light on the molecular mechanisms of the spatial and temporal changes of thousands of cells simultaneously under homeostatic and ischemic conditions. The aim of this study is to investigate whether it is possible to integrate multiple similar scRNA-seq datasets for a more comprehensive understanding of diseases. In this study, we integrated three representative scRNA-seq datasets of 27,349 non-cardiomyocytes isolated at 3 and 7 days after myocardial infarction or sham surgery. In total, seven lineages, including macrophages, fibroblasts, endothelia, and lymphocytes, were identified in this analysis with distinct dynamic and functional properties in healthy and nonhealthy hearts. Myofibroblasts and endothelia were recognized as the central hubs of cellular communication via ligand-receptor interactions. Additionally, we showed that macrophages from different origins exhibited divergent transcriptional signatures, pathways, developmental trajectories, and transcriptional regulons. It was found that myofibroblasts predominantly expand at 7 days after myocardial infarction with pro-reparative characteristics. We identified signature genes of myofibroblasts, such as Postn, Cthrc1, and Ddah1, among which Ddah1 was exclusively expressed on activated fibroblasts and exhibited concordant upregulation in bulk RNA sequencing data and in vivo and in vitro experiments. Collectively, this compendium of scRNA-seq data provides a valuable entry point for understanding the transcriptional and dynamic changes of non-cardiomyocytes in healthy and nonhealthy hearts by integrating multiple datasets.

12.
ESC Heart Fail ; 7(6): 4108-4117, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33006440

RESUMEN

AIMS: In patients with coronavirus disease 2019 (COVID-19), the involvement of the cardiovascular system significantly relates to poor prognosis. However, the risk factors for acute myocardial injury have not been sufficiently studied. Thus, we aimed to determine the characteristics of myocardial injury and define the association between routine blood markers and cardiac troponin I, in order to perform a predictive model. METHODS AND RESULTS: This retrospective cohort study included patients with confirmed COVID-19 from Wuhan Tongji Hospital (Wuhan, China). Data were compared between those with and without myocardial injury. Kaplan-Meier analysis and Cox regression models were used to describe the association between myocardial injury and poor prognosis. Simple correlation analyses were used to find factors associated with high-sensitivity cardiac troponin I levels. Univariate and multivariate logistic regression methods were used to explore the risk factors associated with myocardial injury. The area under the receiver operating characteristic curve was used to determine the predictive value of the model. Of 353 patients included in the study, 79 presented myocardial injury. Patients with myocardial injury had higher levels of inflammation markers, poorer liver and kidney function, and more complications compared with patients without myocardial injury. High-sensitivity cardiac troponin I levels were significantly associated with neutrophil/lymphocyte ratio, creatinine, d-dimer, lactate dehydrogenase, and inflammatory cytokines and negatively associated with oxygen saturation. It was significantly associated with poor prognosis after adjusting for age, sex, and complications. Multivariate regression showed that myocardial injury was associated with a high neutrophil/lymphocyte ratio (odds ratio 2.30, 95% CI 1.11-4.75, per standard deviation increase, P = 0.02), creatinine (3.58, 1.35-8.06, P = 0.01), and lactate dehydrogenase (3.39, 1.42-8.06, P = 0.01) levels. Using a predictive model, the area under the receiver operating characteristic curve was 0.92 (0.88-0.96). CONCLUSIONS: In patients with COVID-19, neutrophil/lymphocyte ratio, creatinine, and lactate dehydrogenase are blood markers that could help identify patients with a high risk of myocardial injury at an early stage.

13.
Am J Physiol Heart Circ Physiol ; 316(5): H971-H984, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30735072

RESUMEN

Fatty acid-binding protein 3 (FABP3), a low-molecular-weight protein, participates in lipid transportation, storage, signaling transduction, oxidation, and transcription regulation. Here, we investigated the expression and function of FABP3 in ischemic heart diseases and explored the mechanisms by which FABP3 affected remodeling after myocardial infarction (MI). We showed that ischemic or hypoxic conditions upregulated FABP3 expression in vivo and in vitro. Notably, overexpression of FABP3 induced more myocyte apoptosis in the infarction and border areas and aggravated cardiac dysfunction, with lower left ventricular ejection fraction. Meanwhile, overexpression of FABP3 drastically promoted death and apoptosis of neonatal rat ventricular cardiomyocytes under hypoxia. Furthermore, deficiency of FABP3 exerted protective effects against ischemic heart injuries by decreasing cardiac myocyte apoptosis and heart remodeling after MI. We found that overexpression of FABP3 upregulated the phosphorylation of MAPK signaling pathway and decreased phosphorylated Akt levels, which may account for the augmentation of apoptosis and remodeling after MI. To the best of our knowledge, this is the first study to demonstrate that deficiency of FABP3 would protect cardiac myocytes from apoptosis and alleviate cardiac remodeling after MI, suggesting FABP3 as a potential target to preserve cardiac function after MI. NEW & NOTEWORTHY It is an undisputable fact that myocyte apoptosis plays a crucial role in cardiac remodeling and the development of heart failure after myocardial infarction. Here, fatty acid-binding protein 3 deficiency improved myocardial structural remodeling and function by decreasing cell apoptosis and regulating MAPK signaling pathways. We suppose that fatty acid-binding protein 3 may be regarded as a potential intervention approach to preserve cardiomyocytes during myocardial infarction.


Asunto(s)
Apoptosis , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 3 de Unión a Ácidos Grasos/deficiencia , Proteína 3 de Unión a Ácidos Grasos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Ratas , Transducción de Señal , Volumen Sistólico , Función Ventricular Izquierda , Remodelación Ventricular
14.
J Cell Mol Med ; 22(10): 4985-4996, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29999589

RESUMEN

Acute kidney injury (AKI) incidence among hospitalized patients is increasing steadily. Despite progress in prevention strategies and support measures, AKI remains correlated with high mortality, particularly among ICU patients, and no effective AKI therapy exists. Here, we investigated the function in kidney ischaemia-reperfusion injury (IRI) of C1orf54, a newly identified protein encoded by an open reading frame on chromosome 1. C1orf54 expression was high in kidney and low in heart, liver, spleen, lung and skeletal muscle in healthy mice, and in the kidney, C1orf54 was expressed in tubular epithelial cells (TECs), but not in glomeruli. C1orf54 expression was markedly decreased on Day 1 after kidney IRI and then gradually recovered to baseline levels by Day 7. Notably, relative to wild-type mice, C1orf54-knockout mice exhibited impaired TEC proliferation and delayed recovery after kidney IRI, which led to deteriorated renal function and increased mortality. Conversely, adenovirus-mediated C1orf54 overexpression promoted TEC proliferation and ameliorated kidney pathology, which resulted in accelerated renal repair and improved renal function. Mechanistically, C1orf54 was found to promote TEC proliferation through PI3K/AKT signalling. Thus, C1orf54 holds considerable potential as a therapeutic target in kidney IRI.


Asunto(s)
Lesión Renal Aguda/genética , Proliferación Celular/genética , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Daño por Reperfusión/genética , Lesión Renal Aguda/patología , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Células Epiteliales/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Daño por Reperfusión/patología , Transducción de Señal/genética
15.
J Int Med Res ; 46(9): 3848-3860, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29962274

RESUMEN

Objective Recent studies have demonstrated that right ventricular apical (RVA) pacing has a deleterious impact on left ventricular function, while right ventricular septum (RVS) or His-bundle pacing (HBP) contribute to improvements in cardiac function. A meta-analysis of randomized controlled trials (RCTs) was conducted to compare the mid- and long-term effects of RVS and HB pacing versus RVA pacing on cardiac function. Methods Eligible RCTs were identified by systematically searching the electronic literature databases PubMed®, Cochrane Library, Embase® and Ovid®. Results Seventeen articles ( n = 1290 patients) were included in this meta-analysis, including 14 studies comparing the effects of RVA and RVS pacing on cardiac function and three studies comparing HBP with pacing at other sites. Compared with RVA pacing, RVS or HBP exhibited a higher left ventricular ejection fraction (LVEF) (weighted mean difference 3.28; 95% confidence interval 1.45, 5.12) at the end of follow-up. Conclusions RVS pacing exhibited a higher LVEF after long-term follow-up than RVA pacing. RVS pacing could replace the previously used RVA pacing as a better alternative with improved clinical outcomes. However, there remains a need for larger RCTs to compare the safety and efficacy of RVS with RVA pacing.


Asunto(s)
Fascículo Atrioventricular , Estimulación Cardíaca Artificial/métodos , Ventrículos Cardíacos , Disfunción Ventricular Izquierda , Función Ventricular/fisiología , Tabique Interventricular , Estimulación Cardíaca Artificial/efectos adversos , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Volumen Sistólico , Resultado del Tratamiento , Disfunción Ventricular/etiología , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA