ZSF1

ZSF1
  • 文章类型: Journal Article
    射血分数保留的心力衰竭(HFpEF)的特征是生物力学功能失调的心肌细胞。潜在的细胞变化包括心肌肌动蛋白表达紊乱和肌动蛋白磷酸化不足,导致肌动蛋白丝变硬。除了这些经过充分研究的心肌细胞水平的改变,运动不耐受是由骨骼肌(SKM)分子改变引起的HFpEF的另一个标志.目前,在HFpEF的SKM中缺乏关于Titin调制的数据。因此,本研究的目的是分析肢体SKM(胫骨前(TA))和diaphragm(Dia)的分子改变,作为一个更中心的SKM,专注于Titin,肌动蛋白磷酸化,和收缩调节蛋白。这项研究是用肌肉组织进行的,从32周龄雌性ZSF-1大鼠获得,建立了HFpEF大鼠模型。我们的结果表明,在肢体SKM中Titin过度磷酸化,基于PEVK区域增强的磷酸化,已知会导致Titin长丝变硬。这种过度磷酸化可以通过高强度间歇训练(HIIT)逆转。此外,肌动蛋白的磷酸化状态与肢体SKM中的肌肉力量之间存在负相关。对于Dia来说,未检测到肌动蛋白磷酸化状态的改变。在以往研究数据的支持下,这表明Dia在HFpEF中具有运动效果。关于收缩调节蛋白的表达,可以检测到Dia和肢体SKM之间的显着差异,支持肢体SKM的肌肉萎缩和功能障碍,但不是在Dia.总之,这些数据表明,在HFpEF中,肌动蛋白硬化与运动不耐受的出现之间存在相关性,以及不同SKM组之间的差异调节。
    Heart failure with preserved ejection fraction (HFpEF) is characterized by biomechanically dysfunctional cardiomyocytes. Underlying cellular changes include perturbed myocardial titin expression and titin hypophosphorylation leading to titin filament stiffening. Beside these well-studied alterations at the cardiomyocyte level, exercise intolerance is another hallmark of HFpEF caused by molecular alterations in skeletal muscle (SKM). Currently, there is a lack of data regarding titin modulation in the SKM of HFpEF. Therefore, the aim of the present study was to analyze molecular alterations in limb SKM (tibialis anterior (TA)) and in the diaphragm (Dia), as a more central SKM, with a focus on titin, titin phosphorylation, and contraction-regulating proteins. This study was performed with muscle tissue, obtained from 32-week old female ZSF-1 rats, an established a HFpEF rat model. Our results showed a hyperphosphorylation of titin in limb SKM, based on enhanced phosphorylation at the PEVK region, which is known to lead to titin filament stiffening. This hyperphosphorylation could be reversed by high-intensity interval training (HIIT). Additionally, a negative correlation occurring between the phosphorylation state of titin and the muscle force in the limb SKM was evident. For the Dia, no alterations in the phosphorylation state of titin could be detected. Supported by data of previous studies, this suggests an exercise effect of the Dia in HFpEF. Regarding the expression of contraction regulating proteins, significant differences between Dia and limb SKM could be detected, supporting muscle atrophy and dysfunction in limb SKM, but not in the Dia. Altogether, these data suggest a correlation between titin stiffening and the appearance of exercise intolerance in HFpEF, as well as a differential regulation between different SKM groups.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由于骨骼肌(SKM)的改变,射血分数保留的心力衰竭(HFpEF)与运动不耐受有关。已知补充亮氨酸会改变合成代谢/分解代谢平衡并改善线粒体功能。因此,我们在已建立的HFpEF大鼠模型中研究了初级和二级预防方法中补充亮氨酸对SKM功能和调节肌肉功能的影响.雌性ZSF1肥胖大鼠被随机分配到未经治疗的,一级预防,二级预防小组。对于一级预防,在HFpEF(8周龄)发病之前开始补充亮氨酸,并进行二级预防,在HFpEF(20周龄)发病后开始补充亮氨酸。在32周龄时评估SKM功能,收集和SKM组织用于线粒体功能评估以及组织学和分子分析。补充亮氨酸可预防SKM功能障碍的发展,但不能逆转。在初级预防组中,线粒体功能改善和mitofilin的更高表达,Mfn-2、Fis1和miCK在SKM中很明显。UCP3的表达减少,而线粒体含量和分解代谢标记(MuRF1,MAFBx),肌肉横截面积,SKM质量没有变化。我们的数据表明,在HFpEF的大鼠模型中,补充亮氨酸可以预防骨骼肌功能障碍的发展。这可能是通过调节能量转移来改善线粒体功能而介导的。
    Heart failure with preserved ejection fraction (HFpEF) is associated with exercise intolerance due to alterations in the skeletal muscle (SKM). Leucine supplementation is known to alter the anabolic/catabolic balance and to improve mitochondrial function. Thus, we investigated the effect of leucine supplementation in both a primary and a secondary prevention approach on SKM function and factors modulating muscle function in an established HFpEF rat model. Female ZSF1 obese rats were randomized to an untreated, a primary prevention, and a secondary prevention group. For primary prevention, leucine supplementation was started before the onset of HFpEF (8 weeks of age) and for secondary prevention, leucine supplementation was started after the onset of HFpEF (20 weeks of age). SKM function was assessed at an age of 32 weeks, and SKM tissue was collected for the assessment of mitochondrial function and histological and molecular analyses. Leucine supplementation prevented the development of SKM dysfunction whereas it could not reverse it. In the primary prevention group, mitochondrial function improved and higher expressions of mitofilin, Mfn-2, Fis1, and miCK were evident in SKM. The expression of UCP3 was reduced whereas the mitochondrial content and markers for catabolism (MuRF1, MAFBx), muscle cross-sectional area, and SKM mass did not change. Our data show that leucine supplementation prevented the development of skeletal muscle dysfunction in a rat model of HFpEF, which may be mediated by improving mitochondrial function through modulating energy transfer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    慢性肾脏病(CKD)进展与持续的氧化应激有关,它通过形成不能被NO激活的氧化且无血红素的apo-sGC来损害NO-sGC-cGMP信号级联。Runcaciguat(BAY1101042)是一部小说,强力,和选择性sGC激活剂,其结合并激活氧化且无血红素的sGC,从而在氧化应激下恢复NO-sGC-cGMP信号传导。因此,runcaciguat可能是CKD/DKD非常有效的治疗选择。在ZSF1大鼠作为CKD/DKD模型中研究了runcaciguat的潜在肾脏保护作用。以高血压为特征,高血糖症,肥胖,和胰岛素抵抗。ZSF1大鼠每天口服治疗长达12周的runcaciguat(1、3、10mg/kg/bid)或安慰剂。研究终点是蛋白尿,肾脏组织病理学,等离子体,肾脏损害的尿液生物标志物,和基因表达谱分析,以获得有关受runcaciguat影响的相关途径的信息。此外,比较了ZSF1大鼠肾脏和DKD患者肾脏样本中的氧化应激。在为期12周的治疗研究期间,肥胖ZSF1大鼠的肾功能显著下降,蛋白尿增加20倍,与瘦ZSF1大鼠相比。Runcaciguat剂量依赖性和显着减弱了ZSF1大鼠蛋白尿的发展,在研究结束时uPCR降低了-19%,-54%,在1、3和10mg/kg/bid时,为-70%,分别,与安慰剂治疗相比。此外,以HbA1C测量的平均血糖水平,甘油三酯,胆固醇增加了五倍,二十次,四次,分别,与瘦大鼠相比,肥胖ZSF1。在肥胖的ZSF1大鼠中,runcaciguat使HbA1c水平降低-8%,-34%,-76%,甘油三酯-42%,-55%,和-71%,胆固醇降低了16%,-17%,-34%,在1、3和10mg/kg/bid下,分别,与安慰剂相比。同时,runcaciguat也减轻了肾脏的重量,肾脏形态学损伤,以及肾脏损害的尿液和血浆生物标志物。有益的作用伴随着基因表达的变化,表明纤维化和炎症减少,并表明内皮稳定性得到改善。总之,在模拟CKD患者常见的合并症和氧化应激条件的DKD大鼠模型中,sGC激活剂runcaciguat显著阻止了肾功能下降.因此,runcaciguat代表CKD患者的一种有希望的治疗选择,这与最近的2期临床研究数据一致,其中runcaciguat在CKD患者中显示出有希望的疗效(NCT04507061)。
    Chronic kidney disease (CKD) progression is associated with persisting oxidative stress, which impairs the NO-sGC-cGMP signaling cascade through the formation of oxidized and heme-free apo-sGC that cannot be activated by NO. Runcaciguat (BAY 1101042) is a novel, potent, and selective sGC activator that binds and activates oxidized and heme-free sGC and thereby restores NO-sGC-cGMP signaling under oxidative stress. Therefore, runcaciguat might represent a very effective treatment option for CKD/DKD. The potential kidney-protective effects of runcaciguat were investigated in ZSF1 rats as a model of CKD/DKD, characterized by hypertension, hyperglycemia, obesity, and insulin resistance. ZSF1 rats were treated daily orally for up to 12 weeks with runcaciguat (1, 3, 10 mg/kg/bid) or placebo. The study endpoints were proteinuria, kidney histopathology, plasma, urinary biomarkers of kidney damage, and gene expression profiling to gain information about relevant pathways affected by runcaciguat. Furthermore, oxidative stress was compared in the ZSF1 rat kidney with kidney samples from DKD patients. Within the duration of the 12-week treatment study, kidney function was significantly decreased in obese ZSF1 rats, indicated by a 20-fold increase in proteinuria, compared to lean ZSF1 rats. Runcaciguat dose-dependently and significantly attenuated the development of proteinuria in ZSF1 rats with reduced uPCR at the end of the study by -19%, -54%, and -70% at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo treatment. Additionally, average blood glucose levels measured as HbA1C, triglycerides, and cholesterol were increased by five times, twenty times, and four times, respectively, in obese ZSF1 compared to lean rats. In obese ZSF1 rats, runcaciguat reduced HbA1c levels by -8%, -34%, and -76%, triglycerides by -42%, -55%, and -71%, and cholesterol by -16%, -17%, and -34%, at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo. Concomitantly, runcaciguat also reduced kidney weights, morphological kidney damage, and urinary and plasma biomarkers of kidney damage. Beneficial effects were accompanied by changes in gene expression that indicate reduced fibrosis and inflammation and suggest improved endothelial stabilization. In summary, the sGC activator runcaciguat significantly prevented a decline in kidney function in a DKD rat model that mimics common comorbidities and conditions of oxidative stress of CKD patients. Thus, runcaciguat represents a promising treatment option for CKD patients, which is in line with recent phase 2 clinical study data, where runcaciguat showed promising efficacy in CKD patients (NCT04507061).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由于其合并症2型糖尿病(T2DM)和高血压,当评估缺血再灌注(IR)损伤时,Zucker自发性高血压脂肪(ZSF1)大鼠是临床相关的动物模型。从糖尿病动物中分离的心脏中的大多数IR研究都是在正常葡萄糖浓度下进行的。提供与体内不同的环境。我们假设在体内调整Krebs缓冲液(KB)时,糖尿病ZSF1大鼠的离体心脏中IR损伤会减弱,即,血糖(BG)水平升高。将糖尿病和非糖尿病ZSF1大鼠麻醉,心孤立和Langendorff准备。标准KB用于非糖尿病和糖尿病未调整组,将葡萄糖水平增加至每只大鼠先前BG水平的KB用于调整的糖尿病组。所有心脏进行30分钟缺血和120分钟再灌注。与未调整的糖尿病和非糖尿病组相比,调整后的缺血和早期再灌注期间的舒张挛缩延迟并暂时减弱。在糖尿病动物中,再灌注时冠状动脉流量的减少减弱。左心室发育压力和收缩力在三组之间没有差异。非糖尿病动物的梗死面积显着降低;缓冲液调整对糖尿病动物没有影响。在我们的研究中,T2DM并没有恶化ZSF1大鼠离体心脏的心肌功能。由于我们的结果表明,调节葡萄糖水平的心脏在IR后显示出至少暂时的功能改善,进一步的研究应该考虑调整葡萄糖水平,以创造更现实的孤立条件,灌注心脏。
    Due to its comorbidities type 2 diabetes mellitus (T2DM) and hypertension, the Zucker Spontaneous Hypertensive Fatty (ZSF1) rat is a clinically relevant animal model when assessing ischemia-reperfusion (IR) injury. Most IR studies in hearts isolated from diabetic animals have been conducted at normal glucose concentrations, providing a different environment compared to in-vivo. We hypothesized IR injury to be attenuated in isolated hearts of diabetic ZSF1 rats when adjusting the Krebs-buffer (KB) to their in-vivo, i.e., elevated blood glucose (BG) levels. Diabetic and non-diabetic ZSF1 rats were anesthetized, hearts isolated and Langendorff-prepared. While standard KB was used for the non-diabetic and diabetic unadjusted groups, KB with glucose levels increased to each rat\'s prior BG level was used for the adjusted diabetic group. All hearts underwent 30 min ischemia and 120 min reperfusion. Diastolic contracture during ischemia and early reperfusion was delayed and temporarily attenuated in the adjusted compared to the unadjusted diabetic and the non-diabetic groups. The decrease in coronary flow on reperfusion was attenuated in diabetic animals. Left ventricular developed pressure and contractility were not different among the three groups. Infarct size was significantly lower in non-diabetic animals; buffer adjustment made no difference in diabetic animals. In our study, T2DM did not worsen myocardial function in ZSF1 rat isolated hearts. Since our results reveal that hearts with an adjusted glucose level exhibit an at least temporary improvement of function following IR, further studies should consider adapting glucose levels to create more realistic conditions in isolated, perfused hearts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    除了心肌的结构改变,射血分数保留的心力衰竭(HFpEF)也与外周骨骼肌(SKM)的分子和生理改变相关,导致HFpEF患者常见的运动不耐受.最近,在临床研究中使用钠-葡萄糖-转运蛋白2抑制剂(SGLT2i)提供了显著降低心血管死亡或HFpEF住院综合风险的证据.本研究旨在进一步阐明Empagliflozin(Empa)对建立的HFpEF大鼠模型中:(1)SKM功能和代谢以及(2)线粒体功能的影响。在24周龄时,肥胖ZSF1大鼠随机接受标准护理或Empa饮用水治疗.ZSF1瘦肉动物作为健康对照。治疗8周后,进行超声心动图和SKM收缩性。在皂苷皮纤维中评估线粒体功能,并将SKM组织快速冷冻以进行分子分析。当与瘦的E/é增加和保留的左心室射血分数相比时,HFpEF在肥胖动物中很明显。Empa治疗显着改善了E/é,并导致SKM收缩力改善,肌内脂质含量降低。在Empa治疗后,线粒体功能更好(主要在复合物IV中),仅对萎缩相关蛋白进行了少量调节。结果清楚地记录了Empa对本发明HFpEF模型中SKM功能的有益作用。这些作用伴随着对线粒体功能的积极作用,可能调节SKM功能。
    Besides structural alterations in the myocardium, heart failure with preserved ejection fraction (HFpEF) is also associated with molecular and physiological alterations of the peripheral skeletal muscles (SKM) contributing to exercise intolerance often seen in HFpEF patients. Recently, the use of Sodium-Glucose-Transporter 2 inhibitors (SGLT2i) in clinical studies provided evidence for a significant reduction in the combined risk of cardiovascular death or hospitalization for HFpEF. The present study aimed to further elucidate the impact of Empagliflozin (Empa) on: (1) SKM function and metabolism and (2) mitochondrial function in an established HFpEF rat model. At the age of 24 weeks, obese ZSF1 rats were randomized either receiving standard care or Empa in the drinking water. ZSF1 lean animals served as healthy controls. After 8 weeks of treatment, echocardiography and SKM contractility were performed. Mitochondrial function was assessed in saponin skinned fibers and SKM tissue was snap frozen for molecular analyses. HFpEF was evident in the obese animals when compared to lean-increased E/é and preserved left ventricular ejection fraction. Empa treatment significantly improved E/é and resulted in improved SKM contractility with reduced intramuscular lipid content. Better mitochondrial function (mainly in complex IV) with only minor modulation of atrophy-related proteins was seen after Empa treatment. The results clearly documented a beneficial effect of Empa on SKM function in the present HFpEF model. These effects were accompanied by positive effects on mitochondrial function possibly modulating SKM function.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    大约一半的心力衰竭(HF)患者,在保留左心室功能的同时,患有舒张功能障碍(所谓的HFpEF)。与可以在药理学上治疗降低的射血分数(HFrEF)的HF相比,对于HFpEF没有特定的治疗剂。心肌肌动蛋白丝硬化,内皮功能障碍,和骨骼肌(SKM)肌病被怀疑有助于HFpEF的发生。我们先前描述了小分子干扰MuRF1靶标识别,从而减轻HFrEF动物模型中的SKM肌病和功能障碍。本研究的目的是测试一种小分子(MyoMed-205)在HFpEF中的功效,并描述MyoMed-205引起的分子变化。
    20周龄的雌性肥胖ZSF1大鼠接受MuRF1抑制剂MyoMed-20512周;与年龄匹配的未经处理的ZSF1瘦(健康)和肥胖大鼠作为对照进行比较。通过超声心动图和侵入性血液动力学测量评估LV(左心室)功能,直到第32周。在第32周,测量SKM和内皮功能并收集组织用于分子分析。应用全蛋白质组分析和WBs和RT-PCR来鉴定特定基因和受影响的分子途径。包括MuRF1敲除小鼠(MuRF1-KO)SKM组织以验证MuRF1特异性。
    到第32周,未经治疗的肥胖大鼠左心室射血分数正常,但E/e比值增加,舒张末期压和心肌纤维化增加,HFpEF的所有典型特征。此外,检测到SKM肌病(萎缩和力丧失)和内皮功能障碍。相比之下,MyoMed-205治疗的大鼠有明显改善的舒张功能,心肌纤维化较少,减少SKM肌病,并增加SKM功能。来自MyoMed-205处理的大鼠的SKM提取物具有降低的MuRF1含量和降低的总肌肉蛋白泛素化。此外,蛋白质组学分析鉴定了八种蛋白质对MyoMed-205治疗有特异性反应。这八种蛋白质中有五种参与线粒体代谢,动力学,或自噬。与线粒体是MyoMed-205靶标一致,在治疗的大鼠中,线粒体呼吸链复合物III的合成增加。MuRF1-KOSKM对照还具有升高的线粒体复合物I和II活性,也表明线粒体活性通过MuRF1调节。
    MyoMed-205在HFpEF的ZSF1动物模型中改善心肌舒张功能并防止SKM萎缩/功能。机械上,SKM受益于减弱的泛素蛋白酶体系统和增强的线粒体呼吸链蛋白质的合成/活性,而心肌似乎受益于减少的肌动蛋白修饰和纤维化。
    About half of heart failure (HF) patients, while having preserved left ventricular function, suffer from diastolic dysfunction (so-called HFpEF). No specific therapeutics are available for HFpEF in contrast to HF where reduced ejection fractions (HFrEF) can be treated pharmacologically. Myocardial titin filament stiffening, endothelial dysfunction, and skeletal muscle (SKM) myopathy are suspected to contribute to HFpEF genesis. We previously described small molecules interfering with MuRF1 target recognition thereby attenuating SKM myopathy and dysfunction in HFrEF animal models. The aim of the present study was to test the efficacy of one small molecule (MyoMed-205) in HFpEF and to describe molecular changes elicited by MyoMed-205.
    Twenty-week-old female obese ZSF1 rats received the MuRF1 inhibitor MyoMed-205 for 12 weeks; a comparison was made to age-matched untreated ZSF1-lean (healthy) and obese rats as controls. LV (left ventricle) function was assessed by echocardiography and by invasive haemodynamic measurements until week 32. At week 32, SKM and endothelial functions were measured and tissues collected for molecular analyses. Proteome-wide analysis followed by WBs and RT-PCR was applied to identify specific genes and affected molecular pathways. MuRF1 knockout mice (MuRF1-KO) SKM tissues were included to validate MuRF1-specificity.
    By week 32, untreated obese rats had normal LV ejection fraction but augmented E/e\' ratios and increased end diastolic pressure and myocardial fibrosis, all typical features of HFpEF. Furthermore, SKM myopathy (both atrophy and force loss) and endothelial dysfunction were detected. In contrast, MyoMed-205 treated rats had markedly improved diastolic function, less myocardial fibrosis, reduced SKM myopathy, and increased SKM function. SKM extracts from MyoMed-205 treated rats had reduced MuRF1 content and lowered total muscle protein ubiquitination. In addition, proteomic profiling identified eight proteins to respond specifically to MyoMed-205 treatment. Five out of these eight proteins are involved in mitochondrial metabolism, dynamics, or autophagy. Consistent with the mitochondria being a MyoMed-205 target, the synthesis of mitochondrial respiratory chain complexes I + II was increased in treated rats. MuRF1-KO SKM controls also had elevated mitochondrial complex I and II activities, also suggesting mitochondrial activity regulation by MuRF1.
    MyoMed-205 improved myocardial diastolic function and prevented SKM atrophy/function in the ZSF1 animal model of HFpEF. Mechanistically, SKM benefited from an attenuated ubiquitin proteasome system and augmented synthesis/activity of proteins of the mitochondrial respiratory chain while the myocardium seemed to benefit from reduced titin modifications and fibrosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Praliciguat, a clinical-stage soluble guanylate cyclase (sGC) stimulator, increases cGMP via the nitric oxide-sGC pathway. Praliciguat has been shown to be renoprotective in rodent models of hypertensive nephropathy and renal fibrosis. In the present study, praliciguat alone and in combination with enalapril attenuated proteinuria in the obese ZSF1 rat model of diabetic nephropathy. Praliciguat monotherapy did not affect hemodynamics. In contrast, enalapril monotherapy lowered blood pressure but did not attenuate proteinuria. Renal expression of genes in pathways involved in inflammation, fibrosis, oxidative stress, and kidney injury was lower in praliciguat-treated obese ZSF1 rats than in obese control rats; fasting glucose and cholesterol were also lower with praliciguat treatment. To gain insight into how tubular mechanisms might contribute to its pharmacological effects on the kidneys, we studied the effects of praliciguat on pathological processes and signaling pathways in cultured human primary renal proximal tubular epithelial cells (RPTCs). Praliciguat inhibited the expression of proinflammatory cytokines and secretion of monocyte chemoattractant protein-1 in tumor necrosis factor-α-challenged RPTCs. Praliciguat treatment also attenuated transforming growth factor-β-mediated apoptosis, changes to a mesenchyme-like cellular phenotype, and phosphorylation of SMAD3 in RPTCs. In conclusion, praliciguat improved proteinuria in the ZSF1 rat model of diabetic nephropathy, and its actions in human RPTCs suggest that tubular effects may contribute to its renal benefits, building upon strong evidence for the role of cGMP signaling in renal health.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Empagliflozin (empa), a selective sodium-glucose cotransporter (SGLT)2 inhibitor, reduced cardiovascular mortality and hospitalization for heart failure in patients with type 2 diabetes at high cardiovascular risk independent of glycemic control. The cardiovascular protective effect of empa was evaluated in an experimental model of metabolic syndrome, the obese ZSF1 rat, and its\' lean control.
    Lean and obese ZSF1 rats were either non-treated or treated with empa (30 mg/kg/day) for 6 weeks. Vascular reactivity was assessed using mesenteric artery rings, systolic blood pressure by tail-cuff sphygmomanometry, heart function and structural changes by echocardiography, and protein expression levels by Western blot analysis.
    Empa treatment reduced blood glucose levels from 275 to 196 mg/dl in obese ZSF1 rats whereas normoglycemia (134 mg/dl) was present in control lean ZSF1 rats and was unaffected by empa. Obese ZSF1 rats showed increased systolic blood pressure, and blunted endothelium-dependent relaxations associated with the appearance of endothelium-dependent contractile responses (EDCFs) compared to control lean rats. These effects were prevented by the empa treatment. Obese ZSF1 rats showed increased weight of the heart and of the left ventricle volume without the presence of diastolic or systolic dysfunction, which were improved by the empa treatment. An increased expression level of senescence markers (p53, p21, p16), tissue factor, VCAM-1, SGLT1 and SGLT2 and a down-regulation of eNOS were observed in the aortic inner curvature compared to the outer one in the control lean rats, which were prevented by the empa treatment. In the obese ZSF1 rats, no such effects were observed. The empa treatment reduced the increased body weight and weight of lungs, spleen, liver and perirenal fat, hyperglycemia and the increased levels of total cholesterol and triglycerides in obese ZSF1 rats, and increased blood ketone levels and urinary glucose excretion in control lean and obese ZSF1 rats.
    Empa reduced glucose levels by 28% and improved both endothelial function and cardiac remodeling in the obese ZSF1 rat. Empa also reduced the increased expression level of senescence, and atherothrombotic markers at arterial sites at risk in the control lean, but not obese, ZSF1 rat.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is underpinned by detrimental skeletal muscle alterations that contribute to disease severity, yet underlying mechanisms and therapeutic treatments remain poorly established. This study used a nonhuman animal model of HFpEF to better understand whether skeletal muscle abnormalities were (1) fiber-type specific and (2) reversible by various exercise training regimes.
    RESULTS: Lean control rats were compared with obese ZSF1 rats at 20 weeks and then 8 weeks after sedentary, high-intensity interval training, or moderate continuous treadmill exercise. Oxidative soleus and glycolytic extensor digitorum longus (EDL) muscles were assessed for fiber size, capillarity, glycolytic metabolism, autophagy, and contractile function. HFpEF reduced fiber size and capillarity by 20%-50% (P < .05) in both soleus and EDL, but these effects were not reversed by endurance training. In contrast, both endurance training regimes in HFpEF attenuated the elevated lactate dehydrogenase activity observed in the soleus. Autophagy was down-regulated in EDL and up-regulated in soleus (P < .05), with no influence of endurance training. HFpEF impaired contractile forces of both muscles by ∼20% (P < .05), and these were not reversed by training.
    CONCLUSIONS: Obesity-related HFpEF was associated with detrimental structural, cellular, and functional alterations to both slow-oxidative and fast-glycolytic skeletal muscles that could not be reversed by endurance training.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:呼吸肌无力导致射血分数(HFpEF)保留的心力衰竭患者的运动不耐受-这种疾病的特征是多种合并症,很少有经过证实的治疗方法。我们的目标是,因此,通过使用肥胖的心脏代谢大鼠模型,提供对HFpEF中潜在的膈肌改变的新见解,并进一步评估仅在明显的HFpEF发展后进行运动训练是否可以逆转损伤。
    结果:肥胖ZSF1大鼠(n=12)与瘦对照(n=8)在20周时进行比较,与另外3组肥胖ZSF1大鼠在8周的久坐行为(n=13)后28周进行比较,高强度间歇训练(n=11),或中度连续训练(n=11)。肥胖大鼠在20和28周时出现明显的HFpEF表型。在20周时的隔膜中,HFpEF诱导了向氧化表型和纤维肥大的转变,同时在MuRF1和MuRF2中蛋白表达降低,但观察到线粒体和收缩功能障碍。28周时,高强度间歇训练或中等强度连续训练的运动训练方案均未逆转HFpEF诱发的膈肌改变.
    结论:这项研究,使用以多种合并症和运动不耐受为基础的特征良好的HFpEF大鼠模型(即,与患者表型非常相似),提供的证据表明,在进行8周的有氧运动训练后,明显的HFpEF引起的diaphragm改变和功能障碍并未逆转。因此,HFpEF患者是否需要其他治疗性干预措施来治疗呼吸肌无力尚需进一步研究.
    BACKGROUND: Respiratory muscle weakness contributes to exercise intolerance in patients with heart failure with a preserved ejection fraction (HFpEF)-a condition characterized by multiple comorbidities with few proven treatments. We aimed, therefore, to provide novel insight into the underlying diaphragmatic alterations that occur in HFpEF by using an obese cardiometabolic rat model and further assessed whether exercise training performed only after the development of overt HFpEF could reverse impairments.
    RESULTS: Obese ZSF1 rats (n=12) were compared with their lean controls (n=8) at 20 weeks, with 3 additional groups of obese ZSF1 rats compared at 28 weeks following 8 weeks of either sedentary behavior (n=13), high-intensity interval training (n=11), or moderate-continuous training (n=11). Obese rats developed an obvious HFpEF phenotype at 20 and 28 weeks. In the diaphragm at 20 weeks, HFpEF induced a shift towards an oxidative phenotype and a fiber hypertrophy paralleled by a lower protein expression in MuRF1 and MuRF2, yet mitochondrial and contractile functional impairments were observed. At 28 weeks, neither the exercise training regimen of high-intensity interval training or moderate-continuous training reversed any of the diaphragm alterations induced by HFpEF.
    CONCLUSIONS: This study, using a well-characterized rat model of HFpEF underpinned by multiple comorbidities and exercise intolerance (ie, one that closely resembles the patient phenotype), provides evidence that diaphragm alterations and dysfunction induced in overt HFpEF are not reversed following 8 weeks of aerobic exercise training. As such, whether alternative therapeutic interventions are required to treat respiratory muscle weakness in HFpEF warrants further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号