AKI-to-CKD transition

AKI 到 CKD 的过渡
  • 文章类型: Journal Article
    急性肾损伤(AKI),以肾功能突然下降为特征,涉及肾小管损伤和上皮细胞死亡,由于间质成纤维细胞活化和缺乏直接治疗的组织修复失败,可导致进行性组织纤维化和慢性肾病。在AKI事件之后,存活的肾小管细胞经历去分化周期,增殖和再分化,而成纤维细胞活性增加,然后下降,以避免过度的细胞外基质沉积。适当的组织恢复与致病性纤维化进展取决于所有这些过程的微调。识别能够影响它们中的任何一个的内源性因素可能为改善AKI结果提供新的治疗机会。半乳糖凝集素-8(Gal-8)是一种内源性碳水化合物结合蛋白,通过非常规机制分泌,与细胞表面的糖基化蛋白质结合并修饰各种细胞活性,包括细胞增殖和在应激条件下的存活。这里,使用叶酸诱导的AKI小鼠模型,我们表明用Gal-8预处理可以防止细胞死亡,促进上皮细胞再分化,改善肾功能。此外,Gal-8减少成纤维细胞活化,导致纤维化基因表达减少。在AKI诱导后添加Gal-8也有效维持肾功能抵抗损伤,提高上皮细胞的存活率。在治疗前后保护肾脏免受损伤的能力,再加上它的抗纤维化作用,强调Gal-8是一种内源性因子,在旨在改善肾功能和缓解慢性致病进展的治疗策略中需要考虑。
    Acute kidney injury (AKI), characterized by a sudden decline in kidney function involving tubular damage and epithelial cell death, can lead to progressive tissue fibrosis and chronic kidney disease due to interstitial fibroblast activation and tissue repair failures that lack direct treatments. After an AKI episode, surviving renal tubular cells undergo cycles of dedifferentiation, proliferation and redifferentiation while fibroblast activity increases and then declines to avoid an exaggerated extracellular matrix deposition. Appropriate tissue recovery versus pathogenic fibrotic progression depends on fine-tuning all these processes. Identifying endogenous factors able to affect any of them may offer new therapeutic opportunities to improve AKI outcomes. Galectin-8 (Gal-8) is an endogenous carbohydrate-binding protein that is secreted through an unconventional mechanism, binds to glycosylated proteins at the cell surface and modifies various cellular activities, including cell proliferation and survival against stress conditions. Here, using a mouse model of AKI induced by folic acid, we show that pre-treatment with Gal-8 protects against cell death, promotes epithelial cell redifferentiation and improves renal function. In addition, Gal-8 decreases fibroblast activation, resulting in less expression of fibrotic genes. Gal-8 added after AKI induction is also effective in maintaining renal function against damage, improving epithelial cell survival. The ability to protect kidneys from injury during both pre- and post-treatments, coupled with its anti-fibrotic effect, highlights Gal-8 as an endogenous factor to be considered in therapeutic strategies aimed at improving renal function and mitigating chronic pathogenic progression.
    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:短链脂肪酸(SCFA),主要是醋酸盐,丙酸和丁酸,由肠道微生物群通过发酵不能被人类宿主消化的复杂碳水化合物产生。它们影响肠道健康,并且可以在远端水平上促进几种疾病的病理生理学,包括肾脏病变。
    方法:在慢性肾脏病(CKD)患者(n=54)的不同疾病阶段测量SCFA水平,并检查其与肾功能和炎症参数的关系。使用全基因组表达测定分析丙酸和丁酸在炎症条件下在肾小管细胞中触发的途径中的影响。最后,叶酸诱导急性肾损伤转化为CKD的临床前小鼠模型用于分析这些微生物代谢产物在CKD发展过程中的预防和治疗潜力.
    结果:CKD患者粪便中丙酸盐和丁酸盐水平随着病情的进展逐渐降低,并与已建立的血清肌酐临床参数密切相关,血尿素氮和估计的肾小球滤过率。丙酸盐和丁酸盐共同下调肾小管细胞中与TNF-α触发的炎症过程和免疫系统激活相关的103个基因的表达。在体内,丙酸和丁酸的管理,在受伤前或受伤后不久,分别防止和减缓损害的发展。肾损伤标志物的减少表明了这一点,促炎和促纤维化标志物的表达,和长期肾功能的恢复。
    结论:丙酸盐和丁酸盐水平与CKD患者肾功能的进行性丧失有关。早期给予这些SCFA可防止急性肾损伤临床前模型中的疾病进展,证明它们的治疗潜力独立于肠道微生物群。
    BACKGROUND: Short-chain fatty acids (SCFAs), mainly acetate, propionate and butyrate, are produced by gut microbiota through fermentation of complex carbohydrates that cannot be digested by the human host. They affect gut health and can contribute at the distal level to the pathophysiology of several diseases, including renal pathologies.
    METHODS: SCFA levels were measured in chronic kidney disease (CKD) patients (n = 54) at different stages of the disease and associations with renal function and inflammation parameters were examined. The impact of propionate and butyrate in pathways triggered in tubular cells under inflammatory conditions was analysed using genome-wide expression assays. Finally, a pre-clinical mouse model of folic acid-induced transition from acute kidney injury to CKD was used to analyse the preventive and therapeutic potential of these microbial metabolites in the development of CKD.
    RESULTS: Faecal levels of propionate and butyrate in CKD patients gradually reduce as the disease progresses, and do so in close association with established clinical parameters for serum creatinine, blood urea nitrogen and the estimated glomerular filtration rate. Propionate and butyrate jointly downregulated the expression of 103 genes related to inflammatory processes and immune system activation triggered by TNF-α in tubular cells. In vivo, the administration of propionate and butyrate, either before or soon after injury, respectively prevented and slowed the progression of damage. This was indicated by a decrease in renal injury markers, the expression of pro-inflammatory and pro-fibrotic markers, and recovery of renal function over the long term.
    CONCLUSIONS: Propionate and butyrate levels are associated with a progressive loss of renal function in CKD patients. Early administration of these SCFAs prevents disease advancement in a pre-clinical model of acute renal damage, demonstrating their therapeutic potential independently of the gut microbiota.
    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
    肠道菌群及其代谢产物会影响全身炎症和肾脏疾病的预后。这里,我们调查了与急性肾损伤(AKI)向慢性肾脏病(CKD)转变相关的关键代谢产物,以及抗生素诱导的微生物群耗竭(AIMD)对这一转变的影响.61例AKI患者中,评估59种血浆代谢物以确定AKI至CKD转变的风险。在单侧缺血再灌注损伤(IRI)四周后建立AKI到CKD转换小鼠模型,以确定AIMD对肠道微生物组的影响。代谢物,以及与CKD转变相关的病理反应。用CKD转换相关代谢物攻击人近端肾小管上皮细胞,并测试了NADPH氧化酶2(NOX2)信号的抑制作用。基于临床代谢组学,血浆氧化三甲胺(TMAO)与AKI至CKD转变风险显著增加相关[校正比值比4.389(95%置信区间1.106~17.416)].在体内,AIMD抑制单侧IRI诱导的TMAO增加,随着细胞凋亡的减少,炎症,和纤维化。AIMD后NOX2的表达和氧化应激降低。体外,TMAO诱导的纤维化具有NOX2激活和氧化应激。NOX2抑制成功减弱细胞凋亡,炎症,和纤维化抑制G2/M阻滞。NOX2抑制(体内)显示病理变化的改善,氧化应激降低,而TMAO水平没有变化。因此,TMAO是与AKI至CKD转换相关的关键代谢产物,并且NOX2激活被鉴定为体内和体外TMAO相关的AKI至CKD转换的关键调节因子。
    Intestinal microbiota and their metabolites affect systemic inflammation and kidney disease outcomes. Here, we investigated the key metabolites associated with the acute kidney injury (AKI)-to chronic kidney disease (CKD) transition and the effect of antibiotic-induced microbiota depletion (AIMD) on this transition. In 61 patients with AKI, 59 plasma metabolites were assessed to determine the risk of AKI-to-CKD transition. An AKI-to-CKD transition murine model was established four weeks after unilateral ischemia-reperfusion injury (IRI) to determine the effects of AIMD on the gut microbiome, metabolites, and pathological responses related to CKD transition. Human proximal tubular epithelial cells were challenged with CKD transition-related metabolites, and inhibitory effects of NADPH oxidase 2 (NOX2) signals were tested. Based on clinical metabolomics, plasma trimethylamine N-oxide (TMAO) was associated with a significantly increased risk for AKI-to-CKD transition [adjusted odds ratio 4.389 (95% confidence interval 1.106-17.416)]. In vivo, AIMD inhibited a unilateral IRI-induced increase in TMAO, along with a decrease in apoptosis, inflammation, and fibrosis. The expression of NOX2 and oxidative stress decreased after AIMD. In vitro, TMAO induced fibrosis with NOX2 activation and oxidative stress. NOX2 inhibition successfully attenuated apoptosis, inflammation, and fibrosis with suppression of G2/M arrest. NOX2 inhibition (in vivo) showed improvement in pathological changes with a decrease in oxidative stress without changes in TMAO levels. Thus, TMAO is a key metabolite associated with the AKI-to-CKD transition, and NOX2 activation was identified as a key regulator of TMAO-related AKI-to-CKD transition both in vivo and in vitro.
    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
    缺氧是肾小管氧化应激和细胞死亡的关键病理生物学触发因素,可驱动急性肾损伤(AKI)向慢性肾脏病(CKD)的转变。富含线粒体的近端肾小管上皮细胞(PTEC)对缺氧非常敏感,因此,在传播AKI到CKD转变的持续肾小管损失方面至关重要。这里,我们研究了PTEC衍生的小细胞外囊泡(sEV)在传播“肾小管死亡波”中的作用。在Transwell插入物上在常氧(21%O2)和缺氧(1%O2)下培养离体患者来源的PTEC,以分离和分析从顶端和基底外侧PTEC表面分泌的sEV。与常氧PTEC相比,低氧PTEC的顶端表面分泌的sEV数量增加。在培养条件之间没有观察到基底外侧sEV数量的差异。低氧根尖sEV货物的生物学途径分析鉴定了与细胞损伤途径相关的不同miRNA。在功能测定中,与常氧根尖sEV相比,低氧根尖sEV选择性诱导自体PTEC中的铁细胞死亡(→谷胱甘肽过氧化物酶-4,→脂质过氧化)。添加铁凋亡抑制剂,铁抑制素-1和黄芩素,减弱的PTEC铁性凋亡。RNA酶A对缺氧根尖sEV的预处理也消除了PTEC铁凋亡,证明sEVRNA在铁细胞死亡波信号传导中的作用。根据这些体外发现,从临床进展为CKD(AKI到CKD转变)的AKI患者的诊断性肾活检的原位免疫标记显示了铁凋亡传播的证据(ACSL4PTEC数量增加),而在功能研究中,来自这些“AKI到CKD转变”患者的尿液来源的sEV(usEV)引发了PTEC铁性凋亡(^脂质过氧化)。我们的数据建立了PTEC衍生的顶端sEV及其囊内RNA作为低氧肾损伤中肾小管脂质过氧化和铁凋亡的介质。肾小管病理学如何从初始损伤传播到“死亡波”的概念为靶向AKI到CKD转变提供了新的治疗检查点。
    Hypoxia is the key pathobiological trigger of tubular oxidative stress and cell death that drives the transition of acute kidney injury (AKI) to chronic kidney disease (CKD). The mitochondrial-rich proximal tubular epithelial cells (PTEC) are uniquely sensitive to hypoxia and thus, are pivotal in propagating the sustained tubular loss of AKI-to-CKD transition. Here, we examined the role of PTEC-derived small extracellular vesicles (sEV) in propagating the \'wave of tubular death\'. Ex vivo patient-derived PTEC were cultured under normoxia (21 % O2) and hypoxia (1 % O2) on Transwell inserts for isolation and analysis of sEV secreted from apical versus basolateral PTEC surfaces. Increased numbers of sEV were secreted from the apical surface of hypoxic PTEC compared with normoxic PTEC. No differences in basolateral sEV numbers were observed between culture conditions. Biological pathway analysis of hypoxic-apical sEV cargo identified distinct miRNAs linked with cellular injury pathways. In functional assays, hypoxic-apical sEV selectively induced ferroptotic cell death (↓glutathione peroxidase-4, ↑lipid peroxidation) in autologous PTEC compared with normoxic-apical sEV. The addition of ferroptosis inhibitors, ferrostatin-1 and baicalein, attenuated PTEC ferroptosis. RNAse A pretreatment of hypoxic-apical sEV also abrogated PTEC ferroptosis, demonstrating a role for sEV RNA in ferroptotic \'wave of death\' signalling. In line with these in vitro findings, in situ immunolabelling of diagnostic kidney biopsies from AKI patients with clinical progression to CKD (AKI-to-CKD transition) showed evidence of ferroptosis propagation (increased numbers of ACSL4+ PTEC), while urine-derived sEV (usEV) from these \'AKI-to-CKD transition\' patients triggered PTEC ferroptosis (↑lipid peroxidation) in functional studies. Our data establish PTEC-derived apical sEV and their intravesicular RNA as mediators of tubular lipid peroxidation and ferroptosis in hypoxic kidney injury. This concept of how tubular pathology is propagated from the initiating insult into a \'wave of death\' provides novel therapeutic check-points for targeting AKI-to-CKD transition.
    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
    急性肾损伤(AKI)后,一些患者的肾功能继续恶化。在促炎和促纤维化的环境中,近端小管会受到适应不良的修复。在AKI到CKD的过渡中,AKI恢复受损会减少肾小管和肾小球滤过,并导致慢性肾脏疾病(CKD)。肾脏分泌能力降低的特征是血浆中生物活性分子的积累,称为尿毒症毒素(UTs)。这些毒素在神经系统的发育中起作用,心血管,骨头,CKD的肾脏并发症。然而,UT也可能导致CKD以及后果。最近的研究表明,这些分子在AKI中早期积累,并有助于在肾脏中建立这种促炎和促纤维化环境。本工作的目的是回顾UT毒性的机制,这些毒性可能导致每个肾室中AKI到CKD的转变。
    After acute kidney injury (AKI), renal function continues to deteriorate in some patients. In a pro-inflammatory and profibrotic environment, the proximal tubules are subject to maladaptive repair. In the AKI-to-CKD transition, impaired recovery from AKI reduces tubular and glomerular filtration and leads to chronic kidney disease (CKD). Reduced kidney secretion capacity is characterized by the plasma accumulation of biologically active molecules, referred to as uremic toxins (UTs). These toxins have a role in the development of neurological, cardiovascular, bone, and renal complications of CKD. However, UTs might also cause CKD as well as be the consequence. Recent studies have shown that these molecules accumulate early in AKI and contribute to the establishment of this pro-inflammatory and profibrotic environment in the kidney. The objective of the present work was to review the mechanisms of UT toxicity that potentially contribute to the AKI-to-CKD transition in each renal compartment.
    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
    急性肾损伤(AKI)影响重症监护病房(ICU)约一半的患者,并恶化他们的短期和长期结果。明显的自限性AKI发作通过细胞和分子机制开始向慢性肾病(CKD)的进展,这些机制尚待解释。特别是,持续性AKI,2016年由急性透析质量倡议定义为AKI,从发病持续48小时以上,与更高的发病率和死亡率相关,与短暂性AKI(即在48小时内逆转的AKI)相比,急性肾病(AKD)和CKD的进展更高。这种分类也被用于实体器官移植的背景中,证明了类似的结果。由于其发病率和预后差,并且由于及时的干预似乎改变了其过程,持续性AKI应及早发现,并在恢复后进行随访。然而,虽然AKI和CKD是很好描述的综合征,持久性AKI和AKD是相对较新的实体。本综述的目的是强调ICU患者持续性AKI在临床和机制特征方面的关键阶段,以便为临床医生和研究人员提供更新的基础,从中开始改善患者的护理和指导未来的研究。
    Acute kidney injury (AKI) affects about half of patients admitted to the intensive care unit (ICU), and worsens their short- and long-term outcomes. Apparently self-limiting AKI episodes initiate a progression toward chronic kidney disease (CKD) through cellular and molecular mechanisms that are yet to be explained. In particular, persistent AKI, defined in 2016 by the Acute Dialysis Quality Initiative as an AKI which lasts more than 48 h from its onset, has been correlated with higher morbidity and mortality, and with a higher progression to acute kidney disease (AKD) and CKD than transient AKI (i.e. AKI with a reversal within 48 h). This classification has been also used in the setting of solid organ transplantation, demonstrating similar outcomes. Due to its incidence and poor prognosis and because prompt interventions seem to change its course, persistent AKI should be recognized early and followed-up also after its recovery. However, while AKI and CKD are well-described syndromes, persistent AKI and AKD are relatively new entities. The purpose of this review is to highlight the key phases of persistent AKI in ICU patients in terms of both clinical and mechanistic features in order to offer to clinicians and researchers an updated basis from which to start improving patients\' care and direct future research.
    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
    急性肾损伤(AKI)和慢性肾病(CKD)是相互关联的综合征,代表着全球公共卫生挑战。这里,我们确定了运动神经元(SMN)存活在缺血/再灌注(I/R)诱导的肾损伤和CKD进展中的特定作用.SMN是所有细胞类型中的必需蛋白,据报道在多个基本细胞稳态途径中起重要作用。然而,SMN在I/R诱导的肾纤维化实验模型中的功能尚未得到广泛研究。SMN的遗传消融或SMN表达的小干扰RNA碱基敲低加重了肾小管损伤和间质纤维化。施用scAAV9-CB-SMN或SMN的上皮细胞过表达减少了I/R诱导的肾功能障碍并减弱了AKI到CKD的转变,表明SMN对肾小管表型的保存和恢复至关重要。我们的数据表明,I/R诱导的内质网应激(ERS)是持续的,并且在没有SMN的肾脏中逐渐变得更加严重。相反,SMN的过表达可预防I/R诱导的ERS和肾小管细胞损伤。总之,我们的数据共同证实了SMN在调节ERS激活和AKI-CKD转换表型方面的关键作用,这可能导致损伤和修复期间的肾脏病理.
    Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected syndromes that represent a global public health challenge. Here, we identified a specific role of survival of motor neuron (SMN) in ischemia/reperfusion (I/R)-induced kidney injury and progression of CKD. SMN was an essential protein in all cell type and was reported to play important roles in multiple fundamental cellular homeostatic pathways. However, the function of SMN in experimental models of I/R-induced kidney fibrosis has not extensively studied. Genetic ablation of SMN or small interfering RNA-base knockdown of SMN expression aggravated the tubular injury and interstitial fibrosis. Administration of scAAV9-CB-SMN or epithelial cell overexpression of SMN reduced I/R-induced kidney dysfunction and attenuated AKI-to-CKD transition, indicating that SMN is vital for the preservation and recovery of tubular phenotype. Our data showed that the endoplasmic reticulum stress (ERS) induced by I/R was persistent and became progressively more severe in the kidney without SMN. On the contrary, overexpression of SMN prevented against I/R-induced ERS and tubular cell damage. In summary, our data collectively substantiate a critical role of SMN in regulating the ERS activation and phenotype of AKI-to-CKD transition that may contribute to renal pathology during injury and repair.
    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
    高迁移率族蛋白盒1(HMGB1)是存在于所有细胞类型中的高度保守的高迁移率族蛋白的成员。HMGB1在细胞内外发挥着多种作用,取决于其亚细胞定位,context,和翻译后修饰。HMGB1还与各种疾病的进展有关。特别是,HMGB1在CKD进展和预后中发挥重要作用。HMGB1通过激活下游信号参与CKD进展中的多个关键事件,包括肾脏炎症,持续性纤维化的发作,肾脏老化,AKI到CKD的过渡,和重要的心血管并发症。更重要的是,HMGB1在肾脏疾病的慢性病理生理学中起着独特的作用。与急性病变不同。这篇综述描述了HMGB1在肾脏稳态中的调节作用,并总结了HMGB1如何影响CKD的进展和预后。最后,总结了靶向抑制HMGB1改善CKD的一些有前景的治疗策略.尽管HMGB1作为治疗靶点在CKD中的应用面临着一些挑战。对HMGB1的细胞内和细胞外调节机制的更深入了解是CKD发生和进展的基础,可能使HMGB1成为CKD有吸引力的治疗靶点.
    High-mobility group protein box 1 (HMGB1) is a member of a highly conserved high-mobility group protein present in all cell types. HMGB1 plays multiple roles both inside and outside the cell, depending on its subcellular localization, context, and post-translational modifications. HMGB1 is also associated with the progression of various diseases. Particularly, HMGB1 plays a critical role in CKD progression and prognosis. HMGB1 participates in multiple key events in CKD progression by activating downstream signals, including renal inflammation, the onset of persistent fibrosis, renal aging, AKI-to-CKD transition, and important cardiovascular complications. More importantly, HMGB1 plays a distinct role in the chronic pathophysiology of kidney disease, which differs from that in acute lesions. This review describes the regulatory role of HMGB1 in renal homeostasis and summarizes how HMGB1 affects CKD progression and prognosis. Finally, some promising therapeutic strategies for the targeted inhibition of HMGB1 in improving CKD are summarized. Although the application of HMGB1 as a therapeutic target in CKD faces some challenges, a more in-depth understanding of the intracellular and extracellular regulatory mechanisms of HMGB1 that underly the occurrence and progression of CKD might render HMGB1 an attractive therapeutic target for CKD.
    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
    急性肾损伤(AKI)后,肾小管上皮细胞(RTEC)的病理特征是细胞内脂滴(LD)积累,参与RTEC损伤和肾纤维化。然而,其发病机制尚不完全清楚。蛋白质,αKlotho,主要以RTEC表示,是众所周知的抗衰老激素,具有多种功能,其膜形式主要充当成纤维细胞生长因子23的共受体。这里,我们发现了膜αKlotho与RTEC细胞内LD之间的联系。荧光脂肪酸(FA)脉冲追踪试验表明,RTEC中膜αKlotho缺乏,如在αKlotho纯合突变(kl/kl)小鼠或缺血再灌注损伤(IRI)诱导的AKI小鼠中所见,通过损害脂肪甘油三酯脂肪酶(ATGL)介导的脂解和吞噬来抑制FA从LD动员。这导致LD积累和FA利用不足。IRI诱导的改变在αKlotho缺乏症中更为明显。机械上,膜αKlotho缺乏促进E3连接酶过氧化物酶2与泛素缀合酶E2D2结合,导致泛素介导的ATGL降解,这是脂解和吞噬的共同分子基础。αKlotho的过表达通过阻止ATGL泛素化来拯救FA动员,从而减少AKI后的LD积累和纤维化。这表明膜αKlotho对于维持RTEC中脂质稳态是必不可少的。因此,我们的研究确定αKlotho是AKI中脂质周转和稳态的关键调节因子,为预防肾小管损伤和AKI向慢性肾脏疾病过渡提供可行的策略。
    After acute kidney injury (AKI), renal tubular epithelial cells (RTECs) are pathologically characterized by intracellular lipid droplet (LD) accumulation, which are involved in RTEC injury and kidney fibrosis. However, its pathogenesis remains incompletely understood. The protein, αKlotho, primarily expressed in RTECs, is well known as an anti-aging hormone wielding versatile functions, and its membrane form predominantly acts as a co-receptor for fibroblast growth factor 23. Here, we discovered a connection between membrane αKlotho and intracellular LDs in RTECs. Fluorescent fatty acid (FA) pulse-chase assays showed that membrane αKlotho deficiency in RTECs, as seen in αKlotho homozygous mutated (kl/kl) mice or in mice with ischemia-reperfusion injury (IRI)-induced AKI, inhibited FA mobilization from LDs by impairing adipose triglyceride lipase (ATGL)-mediated lipolysis and lipophagy. This resulted in LD accumulation and FA underutilization. IRI-induced alterations were more striking in αKlotho deficiency. Mechanistically, membrane αKlotho deficiency promoted E3 ligase peroxin2 binding to ubiquitin-conjugating enzyme E2 D2, resulting in ubiquitin-mediated degradation of ATGL which is a common molecular basis for lipolysis and lipophagy. Overexpression of αKlotho rescued FA mobilization by preventing ATGL ubiquitination, thereby lessening LD accumulation and fibrosis after AKI. This suggests that membrane αKlotho is indispensable for the maintenance of lipid homeostasis in RTECs. Thus, our study identified αKlotho as a critical regulator of lipid turnover and homeostasis in AKI, providing a viable strategy for preventing tubular injury and the AKI-to-chronic kidney disease transition.
    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
    急性肾损伤(AKI)到慢性肾病(CKD)的转变是向终末期肾病缓慢但持续的进展。较早的报道显示,河马组件,例如Yes相关蛋白(YAP)及其同源物TAZ(具有PDZ结合基序的转录共激活因子),在AKI到CKD转变过程中调节炎症和纤维化。值得注意的是,河马组件在AKI过程中的作用和机制各不相同,AKI到CKD的过渡,CKD。因此,详细了解这些角色很重要。这篇综述探讨了Hippo调节因子或成分作为未来治疗目标的潜力,以阻止AKI到CKD的转变。
    Acute kidney injury (AKI)-to-chronic kidney disease (CKD) transition is a slow but persistent progression toward end-stage kidney disease. Earlier reports have shown that Hippo components, such as Yes-associated protein (YAP) and its homolog Transcriptional coactivator with PDZ-binding motif (TAZ), regulate inflammation and fibrogenesis during the AKI-to-CKD transition. Notably, the roles and mechanisms of Hippo components vary during AKI, AKI-to-CKD transition, and CKD. Hence, it is important to understand these roles in detail. This review addresses the potential of Hippo regulators or components as future therapeutic targets for halting the AKI-to-CKD transition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号