Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Med ; 21(1): 96, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36927608

RESUMEN

BACKGROUND: Extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (MSCs) pretreated with atorvastatin (ATV) (MSCATV-EV) have a superior cardiac repair effect on acute myocardial infarction (AMI). The mechanisms, however, have not been fully elucidated. This study aims to explore whether inflammation alleviation of infarct region via macrophage polarization plays a key role in the efficacy of MSCATV-EV. METHODS: MSCATV-EV or MSC-EV were intramyocardially injected 30 min after coronary ligation in AMI rats. Macrophage infiltration and polarization (day 3), cardiac function (days 0, 3, 7, 28), and infarct size (day 28) were measured. EV small RNA sequencing and bioinformatics analysis were conducted for differentially expressed miRNAs between MSCATV-EV and MSC-EV. Macrophages were isolated from rat bone marrow for molecular mechanism analysis. miRNA mimics or inhibitors were transfected into EVs or macrophages to analyze its effects on macrophage polarization and cardiac repair in vitro and in vivo. RESULTS: MSCATV-EV significantly reduced the amount of CD68+ total macrophages and increased CD206+ M2 macrophages of infarct zone on day 3 after AMI compared with MSC-EV group (P < 0.01-0.0001). On day 28, MSCATV-EV much more significantly improved the cardiac function than MSC-EV with the infarct size markedly reduced (P < 0.05-0.0001). In vitro, MSCATV-EV also significantly reduced the protein and mRNA expressions of M1 markers but increased those of M2 markers in lipopolysaccharide-treated macrophages (P < 0.05-0.0001). EV miR-139-3p was identified as a potential cardiac repair factor mediating macrophage polarization. Knockdown of miR-139-3p in MSCATV-EV significantly attenuated while overexpression of it in MSC-EV enhanced the effect on promoting M2 polarization by suppressing downstream signal transducer and activator of transcription 1 (Stat1). Furthermore, MSCATV-EV loaded with miR-139-3p inhibitors decreased while MSC-EV loaded with miR-139-3p mimics increased the expressions of M2 markers and cardioprotective efficacy. CONCLUSIONS: We uncovered a novel mechanism that MSCATV-EV remarkably facilitate cardiac repair in AMI by promoting macrophage polarization via miR-139-3p/Stat1 pathway, which has the great potential for clinical translation.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Infarto del Miocardio , Ratas , Animales , Atorvastatina/farmacología , Atorvastatina/uso terapéutico , Atorvastatina/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Infarto del Miocardio/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Macrófagos/metabolismo , Factor de Transcripción STAT1/metabolismo
2.
Opt Express ; 30(5): 8330-8344, 2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35299576

RESUMEN

Ultra-precision position measurement is increasingly important in advanced manufacturing such as the semiconductor industry and fiber optics or photonics. A vision-based phase estimation method we proposed previously performs position measurement by imaging a 2D periodic pattern. In this paper, systematic errors of this method are analyzed and derived mathematically, which are classified into two types: spectrum leakage error caused by image truncation and window function modulation, and sub-pixel error resulting from discrete Fourier transform (DFT) intensity interpolation. Key design parameters are concluded including pattern period T, camera pixel size t and resolution N, as well as the type of window function used. Numerical simulations are conducted to investigate the relationship between the phase errors and design parameters. Then an error reduction method is proposed. Finally, the improved performance of parameter optimization is validated by a comparative experiment. Experimental results show the measurement errors of the prototype are within ∼2 nm in X or Y axis, and ∼1 µrad in axis, which reaches the sub-pixel accuracy better than 10-3pixel.

3.
Cell Physiol Biochem ; 41(4): 1503-1518, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28334711

RESUMEN

BACKGROUND: The protection of endothelial cells (ECs) against reperfusion injury has received little attention. In this study, we used Tandem Mass Tag (TMT) labeling proteomics to investigate the modulated proteins in an in vitro model of cardiac microvascular endothelial cells (CMECs) subjected to ischemia/reperfusion (I/R) injury and their alteration by traditional Chinese medicine Tongxinluo (TXL). METHODS: Human CMECs were subjected to 2 h of hypoxia followed by 2 h of reoxygenation with different concentrations of TXL Protein expression profiles of CMECs were determined using tandem mass spectrometry. We evaluated several proteins with altered expression in I/R injury and summarized some reported proteins related to I/R injury. RESULTS: TXL dose-dependently decreased CMEC apoptosis, and the optimal concentration was 800 µg/mL. I/R significantly altered proteins in CMECs, and 30 different proteins were detected between a normal group and a hypoxia and serum deprivation group. In I/R injury, TXL treatment up-regulated 6 types of proteins including acyl-coenzyme A synthetase ACSM2B mitochondrial (ACSM2B), cyclin-dependent kinase inhibitor 1B (CDKN1B), heme oxygenase 1 (HMOX1), transcription factor SOX-17 (SOX17), sequestosome-1 isoform 1 (SQSTM1), and TBC1 domain family member 10B (TBC1D10B). Also, TXL down-regulated 5 proteins including angiopoietin-2 isoform c precursor (ANGPT2), cytochrome c oxidase assembly factor 5 (COA5), connective tissue growth factor precursor (CTGF), cathepsin L1 isoform 2 (CTSL), and eukaryotic elongation factor 2 kinase (LOC101930123). These types of proteins mainly had vital functions, including cell proliferation, stress response, and regulation of metabolic process. CONCLUSIONS: The study presented differential proteins upon I/R injury through a proteomic analysis. TXL modulated the expression of proteins in CMECs and has a protective role in response to I/R.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Proteómica , Células Cultivadas , Células Endoteliales/patología , Humanos , Daño por Reperfusión Miocárdica/patología , Miocardio/patología
4.
Chin Med Sci J ; 32(1): 1-12, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28399979

RESUMEN

Objective The benefit of short-term dual antiplatelet therapy (DAPT) following second-generation drug-eluting stents implantation has not been systematically evaluated. To bridge the knowledge gap, we did a meta-analysis to assess the efficacy of ≤6 months versus ≥12 months DAPT among patients with second-generation drug-eluting stents.Methods We searched online databases and identified randomized controlled trials that assess the clinical impact of short-term DAPT (≤6 months) published before March 3, 2016. The efficacy endpoints included the incidence of all-cause death, myocardial infarction, cerebrovascular accidents, and definite or probable stent thrombosis. Safety endpoint defined as major bleeding was also evaluated and discussed.Results We included 5 trials that randomized 9473 participants (49.8%, short-term DAPT duration vs. 50.2%, standard duration). A total of 9445 (99.7%) patients reported the efficacy endpoints, and the safety endpoint was available from 4 studies (n=8457). There was no significant difference in efficacy endpoints between short-term and standard DAPT duration (≥12 months) [risk ratio (RR) 0.96; 95% confidence intervals (CI), 0.80-1.15]. Short-term DAPT duration did not significantly increase the individual risk of all-cause death, myocardial infarction, cerebrovascular accidents, or definite or probable stent thrombosis. Although short-term DAPT obviously reduced risk of major bleeding compared with standard DAPT (RR 0.53; 95% CI, 0.29-0.96), significant publication bias was found when accessing the safety endpoint of the 4 studies (Egger's test, P=0.009).Conclusions The efficacy of short-term DAPT was comparable with that of standard duration DAPT. DAPT less than 6 months may be appropriate for patients receiving second-generation drug-eluting stents implantation.


Asunto(s)
Stents Liberadores de Fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Humanos , Infarto del Miocardio/inducido químicamente , Inhibidores de Agregación Plaquetaria/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Accidente Cerebrovascular/inducido químicamente , Trombosis/inducido químicamente , Factores de Tiempo
5.
Cell Physiol Biochem ; 38(3): 909-25, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26919192

RESUMEN

BACKGROUND/AIMS: Poor viability of transplanted mesenchymal stem cells (MSCs) within the ischemic heart limits their therapeutic potential for cardiac repair. Globular adiponectin (gAPN) exerts anti-apoptotic effects on several types of stem cells. Herein, we investigated the effect of gAPN on the MSCs against apoptosis induced by hypoxia and serum deprivation (H/SD). METHODS: MSCs exposed to H/SD conditions were treated with different concentrations of gAPN. To identify the main type of receptor, MSCs were transfected with siRNA targeting adiponectin receptor 1 or 2 (AdipoR1 or AdipoR2). To elucidate the downstream pathway, MSCs were pre-incubated with AMPK inhibitor Compound C. Apoptosis, caspase-3 activity and mitochondrial membrane potential were evaluated. RESULTS: H/SD-induced MSCs apoptosis and caspase-3 activation were attenuated by gAPN in a concentration-dependent manner. gAPN increased Bcl-2 and decreased Bax expressions. The loss of mitochondrial membrane potential induced by H/SD was also abolished by gAPN. The protective effect of gAPN was significantly attenuated after the knockdown of AdipoR1 rather than AdipoR2. Moreover, Compound C partly suppressed the anti-apoptotic effect of gAPN. CONCLUSIONS: gAPN inhibits H/SD-induced apoptosis in MSCs via AdipoR1-mediated pathway, possibly linked to the activation of AMPK. gAPN may be a novel survival factor for MSCs in the ischemic engraftment environment.


Asunto(s)
Adiponectina/farmacología , Medio de Cultivo Libre de Suero/farmacología , Células Madre Mesenquimatosas/citología , Receptores de Adiponectina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Receptores de Adiponectina/antagonistas & inhibidores
6.
Heart Fail Rev ; 21(6): 737-752, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27459850

RESUMEN

Stem cell therapy is a promising approach to the treatment of ischemic heart disease via replenishing cell loss after myocardial infarction. Both preclinical studies and clinical trials have indicated that cardiac function improved consistently, but very modestly after cell-based therapy. This mainly attributed to low cell survival rate, engraftment and functional integration, which became the major challenges to regenerative medicine. In recent years, several new cell types have been developed to regenerate cardiomyocytes and novel delivery approaches helped to increase local cell retention. New strategies, such as cell pretreatment, gene-based therapy, tissue engineering, extracellular vesicles application and immunologic regulation, have surged and brought about improved cell survival and functional integration leading to better therapeutic effects after cell transplantation. In this review, we summarize these new strategies targeting at challenges of cardiac regenerative medicine and discuss recent evidences that may hint their effectiveness in the future clinical settings.


Asunto(s)
MicroARNs/genética , Isquemia Miocárdica/terapia , Miocitos Cardíacos/citología , Trasplante de Células Madre/métodos , Animales , Regulación de la Expresión Génica , Terapia Genética , Humanos , Ratones , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Regeneración/genética , Ingeniería de Tejidos
7.
Diabetes Res Clin Pract ; 185: 109805, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35219761

RESUMEN

BACKGROUND: Obesity confers paradoxical survival benefits in heart failure with preserved ejection fraction (HFpEF). The purpose of this study was to examine the impact of DM and insulin treatment status on the associations of body mass index (BMI) with the death risks in HFpEF patients. METHODS: HFpEF patients from the TOPCAT trial were included. Cox regression model was constructed to assess the relationship of BMI with the risks of all-cause death and cardiovascular death. Restricted cubic splines were used to characterize the dose-response associations of BMI with risks of death. RESULTS: Compared with normal weight, hazard ratios of all-cause death in overweight and class I obesity were 0.62 (0.45-0.85), 0.67 (0.47-0.94) in no DM HFpEF patients, and 0.48 (0.25-0.91), 0.41 (0.22-0.79) in non-insulin-treated DM patients. However, insulin treatment removed this beneficial effect. Consistent results were found when modeling for time-updated BMI. Cubic spline analyses suggested a linear trend of increased death risk with higher BMI in insulin-treated DM patients. CONCLUSIONS: The "obesity paradox" was present in HFpEF patients without DM or with non-insulin-treated DM but absent in those with insulin-treated DM. Insulin treatment may be a crucial confounder of the obesity paradox in HFpEF patients. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov. Unique identifier: NCT00094302.


Asunto(s)
Diabetes Mellitus , Insuficiencia Cardíaca , Índice de Masa Corporal , Diabetes Mellitus/tratamiento farmacológico , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Insulina/uso terapéutico , Insulina Regular Humana/uso terapéutico , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Volumen Sistólico
8.
J Cardiovasc Transl Res ; 15(3): 514-523, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35229250

RESUMEN

Stem cell-based therapy for ischemic heart disease (IHD) has become a promising but controversial strategy during the past two decades. The fate and effects of stem cells engrafted into ischemia myocardium are still not fully understood. Stem cell-derived exosomes, a subcategory of extracellular vesicles with nano size, have been considered as an efficient and safe transporter for microRNAs (miRNAs) and a central mediator of the cardioprotective potentials of the parental cells. Hypoxia, pharmacological intervention, and gene manipulation could alter the exosomal miRNAs cargos from stem cells and promote therapeutic potential. Furthermore, several bioengineering methods were also successfully applied to modify miRNAs content and components of exosomal membrane proteins recently. In this review, we outline relevant results about exosomal miRNAs from stem cells and focus on the current strategies to promote their therapeutic efficiency in IHD.


Asunto(s)
Exosomas , Vesículas Extracelulares , MicroARNs , Isquemia Miocárdica , Exosomas/genética , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Células Madre/metabolismo
9.
Heart ; 108(17): 1377-1382, 2022 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-35851318

RESUMEN

BACKGROUND: The optimal salt restriction in patients with heart failure (HF), especially patients with heart failure with preserved ejection fraction (HFpEF), remains controversial. OBJECTIVE: To investigate the associations of cooking salt restriction with risks of clinical outcomes in patients with HFpEF. METHODS: Cox proportional hazards model and subdistribution hazards model were used in this secondary analysis in 1713 participants with HFpEF from the Americas in the TOPCAT trial. Cooking salt score was the sum of self-reported salt added during homemade food preparation. The primary endpoint was a composite of cardiovascular death, HF hospitalisation and aborted cardiac arrest, and secondary outcomes were all-cause death, cardiovascular death and HF hospitalisation. RESULTS: Compared with patients with cooking salt score 0, patients with cooking salt score >0 had significantly lower risks of the primary endpoint (HR=0.760, 95% CI 0.638 to 0.906, p=0.002) and HF hospitalisation (HR=0.737, 95% CI 0.603 to 0.900, p=0.003), but not all-cause (HR=0.838, 95% CI 0.684 to 1.027, p=0.088) or cardiovascular death (HR=0.782, 95% CI 0.598 to 1.020, p=0.071). Sensitivity analyses using propensity score matching baseline characteristics and in patients who prepared meals mostly at home yielded similar results. Subgroup analysis suggested that the association between overstrict salt restriction and poor outcomes was more predominant in patients aged ≤70 years and of non-white race. CONCLUSION: Overstrict cooking salt intake restriction was associated with worse prognosis in patients with HFpEF, and the association seemed to be more predominant in younger and non-white patients. Clinicians should be prudent when giving salt restriction advice to patients with HFpEF.


Asunto(s)
Insuficiencia Cardíaca , Humanos , Pronóstico , Modelos de Riesgos Proporcionales , Cloruro de Sodio Dietético/efectos adversos , Volumen Sistólico
10.
Stem Cell Res Ther ; 13(1): 289, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35799283

RESUMEN

BACKGROUND: Bone marrow cells (BMCs), especially mesenchymal stem cells (MSCs), have shown attractive application prospects in acute myocardial infarction (AMI). However, the weak efficacy becomes their main limitation in clinical translation. Based on the anti-inflammation and anti-apoptosis effects of a Chinese medicine-Tongxinluo (TXL), we aimed to explore the effects of TXL-pretreated MSCs (MSCsTXL) in enhancing cardiac repair and further investigated the underlying mechanism. METHODS: MSCsTXL or MSCs and the derived exosomes (MSCsTXL-exo or MSCs-exo) were collected and injected into the infarct zone of rat hearts. In vivo, the anti-apoptotic and anti-inflammation effects, and cardiac functional and histological recovery were evaluated. In vitro, the apoptosis was evaluated by western blotting and flow cytometry. miRNA sequencing was utilized to identify the significant differentially expressed miRNAs between MSCsTXL-exo and MSCs-exo, and the miRNA mimics and inhibitors were applied to explore the specific mechanism. RESULTS: Compared to MSCs, MSCsTXL enhanced cardiac repair with reduced cardiomyocytes apoptosis and inflammation at the early stage of AMI and significantly improved left ventricular ejection fraction (LVEF) with reduced infarct size in an exosome-dependent way. Similarly, MSCsTXL-exo exerted superior therapeutic effects in anti-apoptosis and anti-inflammation, as well as improving LVEF and reducing infarct size compared to MSCs-exo. Further exosomal miRNA analysis demonstrated that miR-146a-5p was the candidate effector of the superior effects of MSCsTXL-exo. Besides, miR-146a-5p targeted and decreased IRAK1, which inhibited the nuclear translocation of NF-κB p65 thus protecting H9C2 cells from hypoxia injury. CONCLUSIONS: This study suggested that MSCsTXL markedly facilitated cardiac repair via a new mechanism of the exosomal transfer of miR-146a-5p targeting IRAK1/NF-κB p65 pathway, which has great potential for clinical translation.


Asunto(s)
Exosomas , Quinasas Asociadas a Receptores de Interleucina-1 , Células Madre Mesenquimatosas , MicroARNs , Infarto del Miocardio , Factor de Transcripción ReIA , Animales , Medicamentos Herbarios Chinos , Exosomas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Ratas , Volumen Sistólico , Factor de Transcripción ReIA/metabolismo , Función Ventricular Izquierda
11.
Stem Cell Res Ther ; 13(1): 63, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35130979

RESUMEN

BACKGROUND: Bone marrow-derived mesenchymal stem cells (MSCs), which possess immunomodulatory characteristic, are promising candidates for the treatment of acute myocardial infarction (AMI). However, the low retention and survival rate of MSCs in the ischemic heart limit their therapeutic efficacy. Strategies either modifying MSCs or alleviating the inflammatory environment, which facilitates the recruitment and survival of the engrafted MSCs, may solve the problem. Thus, we aimed to explore the therapeutic efficacy of sequential transplantation of exosomes and combinatorial pretreated MSCs in the treatment of AMI. METHODS: Exosomes derived from MSCs were delivered to infarcted hearts through intramyocardial injection followed by the intravenous infusion of differentially pretreated MSCs on Day 3 post-AMI. Enzyme linked immunosorbent assay (ELISA) was performed to evaluate the inflammation level as well as the SDF-1 levels in the infarcted border zone of the heart. Echocardiography and histological analysis were performed to assess cardiac function, infarct size, collagen area and angiogenesis. RESULTS: Sequential transplantation of exosomes and the combinatorial pretreated MSCs significantly facilitated cardiac repair compared to AMI rats treated with exosomes alone. Notably, compared to the other three methods of cotransplantation, combinatorial pretreatment with hypoxia and Tongxinluo (TXL) markedly enhanced the CXCR4 level of MSCs and promoted recruitment, which resulted in better cardiac function, smaller infarct size and enhanced angiogenesis. We further demonstrated that exosomes effectively reduced apoptosis in MSCs in vitro. CONCLUSION: Sequential delivery of exosomes and pretreated MSCs facilitated cardiac repair post-AMI, and combined pretreatment with hypoxia and TXL better enhanced the cardioprotective effects. This method provides new insight into the clinical translation of stem cell-based therapy for AMI.


Asunto(s)
Exosomas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Medicamentos Herbarios Chinos , Hipoxia , Trasplante de Células Madre Mesenquimatosas/métodos , Ratas
12.
Mayo Clin Proc ; 97(9): 1619-1630, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36058576

RESUMEN

OBJECTIVE: To define clinical phenotyping and its associated outcome of worsening of renal function (WRF) in hospitalized acute heart failure (AHF) patients. PATIENTS AND METHODS: Latent class analysis was performed in 113 AHF patients who developed WRF within 72 hours in the DOSE (Diuretic Optimization Strategies Evaluation) trial (from March 2008 to November 2009) and ROSE-AHF (Renal Optimization Strategies Evaluation in Acute Heart Failure) trial (from September 2010 to March 2013) to identify potential WRF phenotypes. Clinical characteristics and outcome (in-hospital and post-discharge) were compared between different phenotypes. RESULTS: Two WRF phenotypes were identified by latent class analysis, which we named WRF minimally responsive to diuretics (WRF-MRD) and WRF responsive to diuretics (WRF-RD). Among the population, 58 (9.5%) developed WRF-MRD and 55 (9.0%) developed WRF-RD. Patients with WRF-MRD had more comorbidities than WRF-RD. In WRF-MRD, there were an early increase in serum creatinine, a smaller amount of net fluid loss and weight loss, and a higher rate of worsening or persistent heart failure over 72 hours. In contrast, for those with WRF-RD, they had faster in-hospital net fluid loss and weight loss and a better 60-day survival after discharge even compared with patients without WRF (P=.004). Furthermore, baseline chronic obstructive pulmonary disease, diabetes, and cystatin C were independent predictors of WRF-MRD, whereas serum hemoglobin and sodium predicted WRF-RD. CONCLUSIONS: Among hospitalized AHF patients, we identified two phenotypes of WRF with distinct response to heart failure treatment, predictors, and short-term prognosis after discharge. The results could help early differentiation of WRF phenotypes in clinical practice.


Asunto(s)
Cuidados Posteriores , Insuficiencia Cardíaca , Enfermedad Aguda , Ensayos Clínicos como Asunto , Creatinina , Diuréticos/uso terapéutico , Tasa de Filtración Glomerular/fisiología , Humanos , Riñón/fisiología , Alta del Paciente , Pronóstico , Pérdida de Peso
13.
J Hypertens ; 39(7): 1378-1385, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33534342

RESUMEN

AIMS: To determine the associations of long-term SBP (LT-SBP) levels with clinical outcomes and health-related quality of life in heart failure with preserved ejection fraction (HFpEF). METHODS AND RESULTS: We analyzed participants from the Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist (TOPCAT) study with available different SBP measurements from different follow-ups (n = 3310). LT-SBP was the mean SBP value from 4-week measurement to the last one. The outcome measures are all-cause mortality and a composite of heart failure readmission or all-cause mortality and the Kansas City Cardiomyopathy Questionnaire (KCCQ) overall summary score. To determine the associations of LT-SBP and outcomes, we used adjusted Cox proportional hazards models and restricted cubic spline models. After multivariable adjustment, LT-SBP of 120-129 and 130-139 mmHg were associated with a lower risk of mortality (hazard ratio 0.66, 95% CI 0.51-0.87, P = 0.003; hazard ratio 0.68, 95% CI 0.51-0.90, P = 0.007, respectively); LT-SBP of 100-119 mmHg had similar risk of mortality (hazard ratio 0.96, 95% CI 0.72-1.28, P = 0.778) compared with LT-SBP of at least 140 mmHg. There was U-shaped relationship between LT-SBP and all-cause mortality (P < 0.001) with nadir risk occurring around 123 mmHg. Similar relationships were observed between LT-SBP and composite end point of heart failure readmission or all-cause mortality. The adjusted mean improvement in KCCQ score was significantly higher in the 120-129 mmHg group than in the at least 140 mmHg group beginning from the 12-month follow-up visit without significant differences in other groups. CONCLUSION: Among patients with HFpEF, long-term control of SBP level at 120-129 mmHg is independently associated with the highest risk reduction of all-cause mortality and improvement of KCCQ score. Future randomized clinical trials need to specifically evaluate optimal SBP treatment goals in patients with HFpEF.


Asunto(s)
Insuficiencia Cardíaca , Calidad de Vida , Diuréticos , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Pronóstico , Volumen Sistólico
14.
Front Cardiovasc Med ; 8: 681726, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150872

RESUMEN

Aims: The aim of the study was to determine the associations of weight loss or gain with all-cause mortality risk in heart failure with preserved ejection fraction (HFpEF). Methods and Results: Non-lean patients from the Americas from the Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist study were analyzed (n = 1,515). Weight loss and weight gain were defined as a decrease or increase in weight ≥5% between baseline and 1 year. To determine the associations of weight change and mortality risk, we used adjusted Cox proportional hazards models and restricted cubic spline models. The mean age was 71.5 (9.6) years. Weight loss and gain were witnessed in 19.3 and 15.9% patients, respectively. After multivariable adjustment, weight loss was associated with higher risk of mortality (HR 1.42, 95% CI 1.06-1.89, P = 0.002); weight gain had similar risk of mortality (HR 0.98, 95% CI 0.68-1.42, P = 0.932) compared with weight stability. There was linear relationship between weight change and mortality risk. The association of weight loss and mortality was different for patients with and without diabetes mellitus (interaction p = 0.009). Conclusion: Among patients with HFpEF, weight loss was independently associated with higher risk of all-cause mortality, and weight gain was not associated with better survival. Clinical Trial Registration: https://clinicaltrials.gov, Identifier: NCT00094302.

15.
Front Cardiovasc Med ; 8: 689591, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34195237

RESUMEN

Aims: To investigate the relationship between body-weight fluctuation and risks of clinical outcomes in patients with heart failure with preserved ejection fraction (HFpEF). Methods and Results:We measured intra-individual variations in body weight from baseline and follow-up visits in 1,691 participants with HFpEF from the Americas from the Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist (TOPCAT) trial. The primary endpoint was any cardiovascular events (a composite of death from cardiovascular disease, non-fatal myocardial infarction, aborted cardiac arrest, or hospitalization for HF). The body-weight fluctuation was measured according to average successive variability and high variability was defined as greater than or equal to the median. After adjustment for risk factors, mean body weight and weight change, each increase of 1 standard deviation in body-weight variability was significantly associated with increased risks of any cardiovascular events (hazard ratio [HR] 1.23, 95% confidence interval [CI] 1.15-1.33, P < 0.001). Patients with high variability had a 47% increased risk of any cardiovascular events and 27% increased risk of all-cause death compared with those with low variability. Such association was similar among patients with New York Heart Association functional class I/II vs. III/IV, obesity vs. non-obesity, and weight loss, gain vs. stability (the P-values for interaction were all insignificant). Conclusion: Among patients with HFpEF, body-weight fluctuation was associated with increased risks of cardiovascular events independent of traditional cardiovascular risk factors, and regardless of HF severity, baseline weight or weight change direction. Clinical Trial Registration: Aldosterone antagonist therapy for adults with heart failure and preserved systolic function (TOPCAT), https://clinicaltrials.gov, identifier [NCT00094302].

16.
Cells ; 9(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31979018

RESUMEN

Direct conversion of cardiac fibroblasts into induced cardiomyocytes (iCMs) by forced expression of defined factors holds great potential for regenerative medicine by offering an alternative strategy for treatment of heart disease. Successful iCM conversion can be achieved by minimally using three transcription factors, Mef2c (M), Gata4(G), and Tbx5 (T). Despite increasing interest in iCM mechanistic studies using MGT(polycistronic construct with optimal expression of M,G and T), the reprogramming efficiency varies among different laboratories. Two main Mef2c isoforms (isoform2, Mi2 and isoform4, Mi4) are present in heart and are used separately by different labs, for iCM reprogramming. It is currently unknown if differently spliced isoform of Mef2c contributes to varied reprogramming efficiency. Here, we used Mi2 and Mi4 together with Gata4 and Tbx5 in separate vectors or polycistronic vector, to convert fibroblasts to iCMs. We found that Mi2 can induce higher reprogramming efficiency than Mi4 in MEFs. Addition of Hand2 to MGT retroviral cocktail or polycistronic Mi2-GT retroviruses further enhanced the iCM conversion. Overall, this study demonstrated the isoform specific effects of Mef2c, during iCM reprogramming, clarified some discrepancy about varied efficiency among labs and might lead to future research into the role of alternative splicing and the consequent variants in cell fate determination.


Asunto(s)
Reprogramación Celular , Factores de Transcripción MEF2/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Factores de Transcripción MEF2/química , Ratones Transgénicos , Isoformas de Proteínas/metabolismo
17.
Front Cardiovasc Med ; 7: 592412, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33392270

RESUMEN

Heart failure is the terminal outcome of the majority of cardiovascular diseases, which lacks specific diagnostic biomarkers and therapeutic targets. It contributes to most of cardiovascular hospitalizations and death despite of the current therapy. Therefore, it is important to explore potential molecules improving the diagnosis and treatment of heart failure. MicroRNAs (miRNAs) are small non-coding RNAs that have been reported to be involved in regulating processes of heart failure. After the discovery of miRNAs in exosomes, the subcellular distribution analysis of miRNAs is raising researchers' attention. Growing evidence demonstrates that exosomal miRNAs may be promising diagnostic and therapeutic molecules for heart failure. This review summarizes the role of exosomal miRNAs in heart failure in the prospect of molecular and clinical researches.

18.
Cardiovasc Res ; 116(2): 353-367, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31119268

RESUMEN

AIMS: Naturally secreted nanovesicles, known as exosomes, play important roles in stem cell-mediated cardioprotection. We have previously demonstrated that atorvastatin (ATV) pretreatment improved the cardioprotective effects of mesenchymal stem cells (MSCs) in a rat model of acute myocardial infarction (AMI). The aim of this study was to investigate if exosomes derived from ATV-pretreated MSCs exhibit more potent cardioprotective function in a rat model of AMI and if so to explore the underlying mechanisms. METHODS AND RESULTS: Exosomes were isolated from control MSCs (MSC-Exo) and ATV-pretreated MSCs (MSCATV-Exo) and were then delivered to endothelial cells and cardiomyocytes in vitro under hypoxia and serum deprivation (H/SD) condition or in vivo in an acutely infarcted Sprague-Dawley rat heart. Regulatory genes and pathways activated by ATV pretreatment were explored using genomics approaches and functional studies. In vitro, MSCATV-Exo accelerated migration, tube-like structure formation, and increased survival of endothelial cells but not cardiomyocytes, whereas the exosomes derived from MSCATV-Exo-treated endothelial cells prevented cardiomyocytes from H/SD-induced apoptosis. In a rat AMI model, MSCATV-Exo resulted in improved recovery in cardiac function, further reduction in infarct size and reduced cardiomyocyte apoptosis compared to MSC-Exo. In addition, MSCATV-Exo promoted angiogenesis and inhibited the elevation of IL-6 and TNF-α in the peri-infarct region. Mechanistically, we identified lncRNA H19 as a mediator of the role of MSCATV-Exo in regulating expression of miR-675 and activation of proangiogenic factor VEGF and intercellular adhesion molecule-1. Consistently, the cardioprotective effects of MSCATV-Exo was abrogated when lncRNA H19 was depleted in the ATV-pretreated MSCs and was mimicked by overexpression of lncRNA H19. CONCLUSION: Exosomes obtained from ATV-pretreated MSCs have significantly enhanced therapeutic efficacy for treatment of AMI possibly through promoting endothelial cell function. LncRNA H19 mediates, at least partially, the cardioprotective roles of MSCATV-Exo in promoting angiogenesis.


Asunto(s)
Atorvastatina/farmacología , Células Endoteliales/metabolismo , Exosomas/efectos de los fármacos , Exosomas/trasplante , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Infarto del Miocardio/cirugía , Neovascularización Fisiológica , ARN Largo no Codificante/metabolismo , Proteínas Angiogénicas/metabolismo , Animales , Apoptosis , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Exosomas/metabolismo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética , Ratas Sprague-Dawley , Recuperación de la Función , Regulación hacia Arriba , Función Ventricular Izquierda
19.
Theranostics ; 10(25): 11754-11774, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052245

RESUMEN

Rationale: The crosstalk between cardiac microvascular endothelial cells (CMECs) and cardiomyocytes (CMs) has emerged as a key component in the development of, and protection against, cardiac diseases. For example, activation of endothelial nitric oxide synthase (eNOS) in CMECs, by therapeutic strategies such as ischemic preconditioning, plays a critical role in the protection against myocardial ischemia/reperfusion (I/R) injury. However, much less is known about the signals produced by CMs that are able to regulate CMEC biology. Here we uncovered one such mechanism using Tongxinluo (TXL), a traditional Chinese medicine, that alleviates myocardial ischemia/reperfusion (I/R) injury by activating CMEC eNOS. The aim of our study is to identify the signals produced by CMs that can regulate CMEC biology during I/R. Methods:Ex vivo, in vivo, and in vitro settings of ischemia-reperfusion were used in our study, with the protective signaling pathways activated in CMECs identified using genetic inhibition (p70s6k1 siRNA, miR-145-5p mimics, etc.), chemical inhibitors (the eNOS inhibitor, L-NNA, and the small extracellular vesicles (sEVs) inhibitor, GW4869) and Western blot analyses. TritonX-100 at a dose of 0.125% was utilized to inactivate the eNOS activity in endothelium to investigate the role of CMEC-derived eNOS in TXL-induced cardioprotection. Results: We found that while CMEC-derived eNOS activity was required for the cardioprotection of TXL, activation of eNOS in CMECs by TXL did not occur directly. Instead, eNOS activation in CMECs required a crosstalk between CMs and CMECs through the uptake of CM-derived sEVs. We further demonstrate that TXL induced CM-sEVs contain increased levels of Long Intergenic Non-Protein Coding RNA, Regulator Of Reprogramming (Linc-ROR). Upon uptake into CMECs, linc-ROR downregulates its target miR-145-5p leading to activation of the eNOS pathway by facilitating the expression of p70s6k1 in these cells. The activation of CMEC-derived eNOS works to increase survival in both the CMECs and the CMs themselves. Conclusions: These data uncover a mechanism by which the crosstalk between CMs and CMECs leads to the increased survival of the heart after I/R injury and point to a new therapeutic target for the blunting of myocardial I/R injury.


Asunto(s)
Cardiotónicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Cardiotónicos/uso terapéutico , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/citología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Humanos , Preparación de Corazón Aislado , Masculino , Microvasos/citología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Nitroarginina/farmacología , Ratas , Transducción de Señal/efectos de los fármacos
20.
Sci Transl Med ; 12(566)2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087505

RESUMEN

Direct reprogramming of fibroblasts to alternative cell fates by forced expression of transcription factors offers a platform to explore fundamental molecular events governing cell fate identity. The discovery and study of induced cardiomyocytes (iCMs) not only provides alternative therapeutic strategies for heart disease but also sheds lights on basic biology underlying CM fate determination. The iCM field has primarily focused on early transcriptome and epigenome repatterning, whereas little is known about how reprogramming iCMs remodel, erase, and exit the initial fibroblast lineage to acquire final cell identity. Here, we show that autophagy-related 5 (Atg5)-dependent autophagy, an evolutionarily conserved self-digestion process, was induced and required for iCM reprogramming. Unexpectedly, the autophagic factor Beclin1 (Becn1) was found to suppress iCM induction in an autophagy-independent manner. Depletion of Becn1 resulted in improved iCM induction from both murine and human fibroblasts. In a mouse genetic model, Becn1 haploinsufficiency further enhanced reprogramming factor-mediated heart function recovery and scar size reduction after myocardial infarction. Mechanistically, loss of Becn1 up-regulated Lef1 and down-regulated Wnt inhibitors, leading to activation of the canonical Wnt/ß-catenin signaling pathway. In addition, Becn1 physically interacts with other classical class III phosphatidylinositol 3-kinase (PI3K III) complex components, the knockdown of which phenocopied Becn1 depletion in cardiac reprogramming. Collectively, our study revealed an inductive role of Atg5-dependent autophagy as well as a previously unrecognized autophagy-independent inhibitory function of Becn1 in iCM reprogramming.


Asunto(s)
Reprogramación Celular , Fosfatidilinositol 3-Quinasas , Animales , Autofagia , Beclina-1/metabolismo , Regulación hacia Abajo , Fibroblastos/metabolismo , Ratones , Miocitos Cardíacos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA