proximal tubule

近端小管
  • 文章类型: Journal Article
    粘菌素是一种多粘菌素抗生素,由于其在治疗多药耐药(MDR)细菌感染中的功效,目前正在引起新的临床兴趣。急性剂量依赖性肾损伤的频繁发作,有可能导致长期的肾损伤,限制了其使用并阻碍了适当的给药方案,增加治疗期间血浆浓度次优的风险。粘菌素诱导的肾毒性机制已被假定为源于线粒体损伤,然而,没有直接的证据表明粘菌素作为线粒体毒素。这项研究的目的是评估粘菌素是否可以直接诱导线粒体毒性,如果是,揭示潜在的分子机制。我们发现粘菌素导致从小鼠肾脏分离的线粒体的快速通透性转变,这通过线粒体与线粒体过渡孔环孢菌素A或L-肉碱的脱敏剂的共孵育而被完全阻止。在原代培养的小鼠肾小管细胞的实验中证实了L-肉碱的保护作用。始终如一,粘菌素诱导的肾脏损害的相对风险,根据组织学分析以及肾小管损伤的早期标志物计算,Kim-1在体内与L-肉碱共同施用下减半。值得注意的是,左旋肉碱既不影响粘菌素的药代动力学,也不影响其对相关细菌菌株的抗菌活性。总之,粘菌素靶向线粒体并诱导其通透性转变。左旋肉碱在体外预防粘菌素诱导的通透性转变。此外,左卡尼汀共同给药赋予用粘菌素治疗的小鼠部分肾保护作用,而不会干扰其药代动力学和抗菌活性。
    Colistin is a polymyxin antibiotic currently experiencing renewed clinical interest due to its efficacy in the treatment of multidrug resistant (MDR) bacterial infections. The frequent onset of acute dose-dependent kidney injury, with the potential of leading to long-term renal damage, has limited its use and hampered adequate dosing regimens, increasing the risk of suboptimal plasma concentrations during treatment. The mechanism of colistin-induced renal toxicity has been postulated to stem from mitochondrial damage, yet there is no direct evidence of colistin acting as a mitochondrial toxin. The aim of this study was to evaluate whether colistin can directly induce mitochondrial toxicity and, if so, uncover the underlying molecular mechanism. We found that colistin leads to a rapid permeability transition of mitochondria isolated from mouse kidney that was fully prevented by co-incubation of the mitochondria with desensitizers of the mitochondrial transition pore cyclosporin A or L-carnitine. The protective effect of L-carnitine was confirmed in experiments in primary cultured mouse tubular cells. Consistently, the relative risk of colistin-induced kidney damage, calculated based on histological analysis as well as by the early marker of tubular kidney injury, Kim-1, was halved under co-administration with L-carnitine in vivo. Notably, L-carnitine neither affected the pharmacokinetics of colistin nor its antimicrobial activity against relevant bacterial strains. In conclusion, colistin targets the mitochondria and induces permeability transition thereof. L-carnitine prevents colistin-induced permeability transition in vitro. Moreover, L-carnitine co-administration confers partial nephroprotection in mice treated with colistin, without interfering with its pharmacokinetics and antibacterial activity.
    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
    几种药物引起的直接肾小管损伤,尤其是化疗药物,是AKI的常见原因。细胞周期蛋白依赖性激酶12(CDK12)的抑制或缺失引发转录延伸缺陷,导致DNA损伤修复缺陷,在各种癌症中产生基因组不稳定性。值得注意的是,10-25%的人在用CDK12抑制剂治疗后发生AKI,潜在的机制还不清楚。这里,我们发现,在AKI患者和小鼠AKI模型中,CDK12在肾小管上皮细胞中下调.此外,小鼠中CDK12的肾小管细胞特异性敲低通过促进基因组不稳定性增强顺铂诱导的AKI,凋亡,和增殖抑制,而CDK12过表达保护抗AKI。在CDK12RTEC+/-小鼠的肾脏上使用单分子实时(SMRT)平台,我们发现CDK12敲低通过转录延伸缺陷靶向Fgf1和Cast,从而增强基因组的不稳定性和细胞凋亡。总的来说,这些数据表明,CDK12敲低可以通过改变Fgf1和Cast基因的转录延伸缺陷来增强AKI的发展,对CDK12抑制剂治疗的患者应给予更多的关注以预防AKI。
    Direct tubular injury caused by several medications, especially chemotherapeutic drugs, is a common cause of AKI. Inhibition or loss of cyclin-dependent kinase 12 (CDK12) triggers a transcriptional elongation defect that results in deficiencies in DNA damage repair, producing genomic instability in a variety of cancers. Notably, 10-25% of individuals developed AKI after treatment with a CDK12 inhibitor, and the potential mechanism is not well understood. Here, we found that CDK12 was downregulated in the renal tubular epithelial cells in both patients with AKI and murine AKI models. Moreover, tubular cell-specific knockdown of CDK12 in mice enhanced cisplatin-induced AKI through promotion of genome instability, apoptosis, and proliferative inhibition, whereas CDK12 overexpression protected against AKI. Using the single molecule real-time (SMRT) platform on the kidneys of CDK12RTEC+/- mice, we found that CDK12 knockdown targeted Fgf1 and Cast through transcriptional elongation defects, thereby enhancing genome instability and apoptosis. Overall, these data demonstrated that CDK12 knockdown could potentiate the development of AKI by altering the transcriptional elongation defect of the Fgf1 and Cast genes, and more attention should be given to patients treated with CDK12 inhibitors to prevent AKI.
    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
    目的:观察烟酰胺(Nam)对糖尿病肾病(DKD)小鼠的影响并探讨其作用机制。
    方法:30只DBA/2J小鼠随机分为三组。链脲佐菌素(STZ)诱导高血糖8周后,对STZ+Nam和STZ+NS小鼠给予Nam和生理盐水,分别,八个星期。非糖尿病小鼠(NDM)作为对照组。将20只In2-/-秋田小鼠随机分为两组。高血糖8周后,对Akita+Nam和Akita+NS小鼠给予Nam和生理盐水,分别,6周。野生型同窝作为对照组。分析肾损伤的标志物,并探讨了人近端肾小管HK2细胞的分子机制。
    结果:STZ+Nam和Akita+Nam组的尿白蛋白-肌酐比值(UACR)和肾损伤分子1(KIM-1)降低。病理分析显示Nam改善了肾小球基底膜的结构,改善的肾小球硬化,减少了细胞外基质和胶原的积累。与糖尿病对照组相比,肾纤维化,炎症,在Nam处理的小鼠中,氧化应激降低。高糖抑制了人近端肾小管HK2细胞中沉默调节蛋白1(Sirt1)的表达,而Nam处理增强了其表达。然而,在具有Sirt1敲低的HK2细胞中,Nam的保护作用被废除了,表明Nam的有益作用部分依赖于Sirt1。
    结论:Nam对高血糖引起的肾损伤具有肾脏保护作用,可能是治疗DKD的潜在靶点。
    OBJECTIVE: To investigate the effect of nicotinamide (Nam) on diabetic kidney disease (DKD) in mice and explore its mechanism.
    METHODS: Thirty DBA/2 J mice were randomly assigned to three groups. After 8 weeks of hyperglycemia induced by streptozocin (STZ), Nam and saline were administrated to STZ + Nam and STZ + NS mice, respectively, for 8 weeks. Non-diabetic mice (NDM) were used as control group. Twenty In2-/- Akita mice were randomly divided into two groups. After 8 weeks of hyperglycemia, Nam and saline were administered to Akita + Nam and Akita + NS mice, respectively, for 6 weeks. Wild-type littermates were used as control group. Markers of renal injury were analyzed, and the molecular mechanisms were explored in human proximal tubular HK2 cells.
    RESULTS: Urinary albumin-to-creatinine ratio (UACR) and kidney injury molecule 1 (KIM-1) decreased in the STZ + Nam and Akita + Nam groups. Pathological analysis showed that Nam improved the structure of glomerular basement membrane, ameliorated glomerular sclerosis, and decreased the accumulation of extracellular matrix and collagen. Compared to the diabetic control group, renal fibrosis, inflammation, and oxidative stress were reduced in the Nam-treated mice. The expression of sirtuin 1 (Sirt1) in human proximal tubular HK2 cells was inhibited by high glucose and Nam treatment enhanced its expression. However, in HK2 cells with Sirt1 knockdown, the protective effect of Nam was abolished, indicating that the beneficial effect of Nam was partially dependent on Sirt1.
    CONCLUSIONS: Nam has a renoprotective effect against renal injury caused by hyperglycemia and may be a potential target for the treatment of DKD.
    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
    背景:基底外侧钾通道在维持肾脏近端小管(PT)的膜运输中起重要作用,腺苷受体已被证明可以调节PT中的跨上皮Na吸收。本研究的目的是探讨腺苷是否也调节PT的基底外侧K通道,并确定腺苷受体类型和介导腺苷对K通道作用的信号通路。方法:我们使用单通道记录来检查小鼠肾脏近端小管的基底外侧K通道活性。所有实验均在细胞附着的贴片中进行。结果:单通道记录已检测到具有高通道开放概率的50pS向内整流K通道,并且该50pSK通道是小鼠PT基底外侧膜中的主要K型通道。在细胞附着的贴片中,添加腺苷增加了50pSK+通道活性,由NPo(通道号和开放概率的乘积)定义。在用DPCPX预处理的PT中不存在腺苷诱导的50pSK通道刺激,腺苷A1受体的选择性抑制剂。相比之下,腺苷仍然能够刺激用选择性腺苷A2受体拮抗剂CP-66713预处理的PT中的50pSK通道。这表明腺苷对PT的50pSK通道的刺激作用是由腺苷A1受体介导的。此外,在用U-73122或CalphostinC预处理的PT中,腺苷对50pSK通道的作用被阻断,表明腺苷诱导的PT的50pSK通道的刺激是由于磷脂酶C(PLC)和蛋白激酶C(PKC)途径的激活。相比之下,AACOCF3抑制磷脂酶A2(PLA2)或H8抑制蛋白激酶A(PKA)未能阻断腺苷诱导的PT50pSK通道的刺激。结论:我们得出结论,腺苷通过腺苷A1受体激活PT基底外侧膜中的50pSK通道。此外,腺苷对50pSK+通道的作用是通过PLC-PKC信号通路介导的。
    Background: The basolateral potassium channels play an important role in maintaining the membrane transport in the renal proximal tubules (PT) and adenosine receptors have been shown to regulate the trans-epithelial Na+ absorption in the PT. The aim of the present study is to explore whether adenosine also regulates the basolateral K+ channel of the PT and to determine the adenosine receptor type and the signaling pathway which mediates the effect of adenosine on the K+ channel. Methods: We have used the single channel recording to examine the basolateral K+ channel activity in the proximal tubules of the mouse kidney. All experiments were performed in cell-attached patches. Results: Single channel recording has detected a 50 pS inwardly-rectifying K+ channel with high channel open probability and this 50 pS K+ channel is a predominant type K+ channel in the basolateral membrane of the mouse PT. Adding adenosine increased 50 pS K+ channel activity in cell-attached patches, defined by NPo (a product of channel Numbers and Open Probability). The adenosine-induced stimulation of the 50 pS K+ channel was absent in the PT pretreated with DPCPX, a selective inhibitor of adenosine A1 receptor. In contrast, adenosine was still able to stimulate the 50 pS K+ channel in the PT pretreated with CP-66713, a selective adenosine A2 receptor antagonist. This suggests that the stimulatory effect of adenosine on the 50 pS K+ channel of the PT was mediated by adenosine-A1 receptor. Moreover, the effect of adenosine on the 50 pS K+ channel was blocked in the PT pretreated with U-73122 or Calphostin C, suggesting that adenosine-induced stimulation of the 50 pS K+ channels of the PT was due to the activation of phospholipase C (PLC) and protein kinase C (PKC) pathway. In contrast, the inhibition of phospholipase A2 (PLA2) with AACOCF3 or inhibition of protein kinase A (PKA) with H8 failed to block the adenosine-induced stimulation of the 50 pS K+ channel of the PT. Conclusion: We conclude that adenosine activates the 50 pS K+ channels in the basolateral membrane of PT via adenosine-A1 receptor. Furthermore, the effect of adenosine on the 50 pS K+ channel is mediated by PLC-PKC signaling pathway.
    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)是导致肾脏纤维化的关键临床疾病,目前缺乏具体的治疗方案。在这项研究中,我们使用叶酸(FA)和缺血再灌注(IRI)诱导的AKI模型,研究了SENP1-Sirt3信号通路的作用及其与近端肾小管上皮细胞(PTEC)线粒体功能障碍的相关性.我们的发现表明,遗传点突变(Sirt3KR)或药物刺激(二甲双胍)通过降低线粒体SOD2的乙酰化水平,减少线粒体ROS(mtROS)来保护小鼠免受AKI和随后的肾脏炎症和纤维化,随后恢复线粒体ATP水平,逆转线粒体形态,减轻细胞凋亡。此外,通过使用N-乙酰基-L-半胱氨酸(NAC)或MitoQ降低mtROS水平,小鼠中的AKI得到类似缓解。代谢组学分析进一步表明,在AKI期间,Sirt3KR小鼠的抗氧化剂和代谢变化增加,与Sirt3WT小鼠相比。使用二甲双胍激活AMPK途径促进SENP1-Sirt3轴并保护PTEC免于凋亡。因此,线粒体中Sirt3的增强去SUMO化,通过代谢相关的AMPK途径激活,通过Sirt3-SOD2-mtROS保护AKI并随后减轻肾脏炎症和纤维化,这代表了AKI的潜在治疗靶标。
    Acute kidney injury (AKI) is a critical clinical condition that causes kidney fibrosis, and it currently lacks specific treatment options. In this research, we investigate the role of the SENP1-Sirt3 signaling pathway and its correlation with mitochondrial dysfunction in proximal tubular epithelial cells (PTECs) using folic acid (FA) and ischemia-reperfusion-induced (IRI) AKI models. Our findings reveal that Sirt3 SUMOylation site mutation (Sirt3 KR) or pharmacological stimulation (metformin) protected mice against AKI and subsequent kidney inflammation and fibrosis by decreasing the acetylation level of mitochondrial SOD2, reducing mitochondrial reactive oxygen species (mtROS), and subsequently restoring mitochondrial ATP level, reversing mitochondrial morphology and alleviating cell apoptosis. In addition, AKI in mice was similarly alleviated by reducing mtROS levels using N-acetyl-L-cysteine (NAC) or MitoQ. Metabolomics analysis further demonstrated an increase in antioxidants and metabolic shifts in Sirt3 KR mice during AKI, compared with Sirt3 wild-type (WT) mice. Activation of the AMPK pathway using metformin promoted the SENP1-Sirt3 axis and protected PTECs from apoptosis. Hence, the augmented deSUMOylation of Sirt3 in mitochondria, activated through the metabolism-related AMPK pathway, protects against AKI and subsequently mitigated renal inflammation and fibrosis through Sirt3-SOD2-mtROS, which represents a potential therapeutic target for AKI.
    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
    肾小管间质纤维化(TIF),几乎所有进行性慢性肾脏疾病(CKD)的共同最终结果,也是肾脏存活的最佳预测因子。肾脏中几乎所有的细胞都参与了TIF的进展。肌成纤维细胞,细胞外基质的初级生产者,以前受到了很多关注;然而,大量新出现的证据表明,近端小管(PT)在TIF进展中起着重要作用。为了应对伤害,肾小管上皮细胞(TECs)转化为炎症和成纤维细胞,产生驱动间质炎症和纤维化的各种生物活性分子。在这里,我们回顾了越来越多的证据,表明PT在促进TIF在肾小管间质和肾小球损伤中的关键作用,并讨论了涉及PT的治疗靶标和载体系统,这对于治疗纤维化肾病患者具有特别的希望。
    Tubulointerstitial fibrosis (TIF), a common end result of almost all progressive chronic kidney diseases (CKD), is also the best predictor of kidney survival. Almost all cells in the kidney are involved in the progression of TIF. Myofibroblasts, the primary producers of extracellular matrix, have previously received a great deal of attention; however, a large body of emerging evidence reveals that proximal tubule (PT) plays a central role in TIF progression. In response to injury, renal tubular epithelial cells (TECs) transform into inflammatory and fibroblastic cells, producing various bioactive molecules that drive interstitial inflammation and fibrosis. Here we reviewed the increasing evidence for the key role of the PT in promoting TIF in tubulointerstitial and glomerular injury and discussed the therapeutic targets and carrier systems involving the PT that holds particular promise for treating patients with fibrotic nephropathy.
    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,肾小管细胞可以刺激成纤维细胞的旁分泌方式导致间质纤维化,但是在这种情况下,旁分泌因子及其调节仍然难以捉摸。在这里,我们确定了巨自噬/自噬依赖性FGF2(成纤维细胞生长因子2)在肾小管细胞中的生产。诱导后,FGF2作为激活肾纤维化的成纤维细胞的关键旁分泌因子。小鼠缺血性AKI后,自噬激活在肾小管细胞中持续数周。在诱导型中,肾小管特异性atg7(自噬相关7)敲除(iRT-atg7-KO)小鼠,AKI后诱导的自噬缺陷抑制了肾小管细胞的促纤维化表型并减少了纤维化。在主要的细胞因子中,iRT-atg7-KO小鼠的肾小管自噬缺陷特异性减少FGF2。自噬抑制也减弱了TGFB1/TGF-β1中FGF2的表达(转化生长因子,β1)处理的肾小管细胞。与旁分泌作用一致,TGFB1处理的肾小管细胞的培养基刺激肾成纤维细胞,并且这种作用被FGF2中和抗体以及肾小管细胞中的fgf2-或atg7-缺失抑制。在人类中,与非AKI相比,AKI后患者的肾活检在肾小管细胞中具有较高水平的自噬和FGF2,与肾脏纤维化呈显著相关性。这些结果表明,AKI后持续的自噬诱导肾小管细胞中的促纤维化表型转化,导致FGF2的表达和分泌,这在适应不良的肾脏修复期间激活了肾纤维化的成纤维细胞。缩写:3-MA:3-甲基腺苷;ACTA2/α-SMA:肌动蛋白α2,平滑肌,主动脉;ACTB/β-肌动蛋白:肌动蛋白,β;AKI:急性肾损伤;ATG/Atg:自噬相关;BUN:血尿素氮;CCN2/CTGF:细胞通讯网络因子2;CDKN2A/p16:细胞周期蛋白依赖性激酶抑制剂2A;CKD:慢性肾病;CM:条件培养基;COL1A1:胶原,I型,α1;COL4A1:胶原蛋白,IV型,α1;CQ:氯喹;ECM:细胞外基质;eGFR:估计的肾小球滤过率;ELISA:酶联免疫吸附测定;FGF2:成纤维细胞生长因子2;FN1:纤连蛋白1;FOXO3:叉头盒O3;GAPDH:甘油醛-3-磷酸脱氢酶;HAVCR1/KIM-1:甲型肝炎病毒细胞受体1;IHC:四联免疫组织化学:四联蛋白;TB多肽;PPIB/亲环蛋白B:肽基氨酰异构酶B;RT-qPCR:实时定量PCR;SA-GLB1/β-gal:衰老相关半乳糖苷酶,β1;SASP:衰老相关分泌表型;sCr:血清肌酐;SQSTM1/p62:隔离体1;TASCC:TOR-自噬空间耦合区室;TGFB1/TGF-β1:转化生长因子,β1;VIM:波形蛋白。
    Following acute kidney injury (AKI), renal tubular cells may stimulate fibroblasts in a paracrine fashion leading to interstitial fibrosis, but the paracrine factors and their regulation under this condition remain elusive. Here we identify a macroautophagy/autophagy-dependent FGF2 (fibroblast growth factor 2) production in tubular cells. Upon induction, FGF2 acts as a key paracrine factor to activate fibroblasts for renal fibrosis. After ischemic AKI in mice, autophagy activation persisted for weeks in renal tubular cells. In inducible, renal tubule-specific atg7 (autophagy related 7) knockout (iRT-atg7-KO) mice, autophagy deficiency induced after AKI suppressed the pro-fibrotic phenotype in tubular cells and reduced fibrosis. Among the major cytokines, tubular autophagy deficiency in iRT-atg7-KO mice specifically diminished FGF2. Autophagy inhibition also attenuated FGF2 expression in TGFB1/TGF-β1 (transforming growth factor, beta 1)-treated renal tubular cells. Consistent with a paracrine action, the culture medium of TGFB1-treated tubular cells stimulated renal fibroblasts, and this effect was suppressed by FGF2 neutralizing antibody and also by fgf2- or atg7-deletion in tubular cells. In human, compared with non-AKI, the renal biopsies from post-AKI patients had higher levels of autophagy and FGF2 in tubular cells, which showed significant correlations with renal fibrosis. These results indicate that persistent autophagy after AKI induces pro-fibrotic phenotype transformation in tubular cells leading to the expression and secretion of FGF2, which activates fibroblasts for renal fibrosis during maladaptive kidney repair.Abbreviations: 3-MA: 3-methyladnine; ACTA2/α-SMA: actin alpha 2, smooth muscle, aorta; ACTB/β-actin: actin, beta; AKI: acute kidney injury; ATG/Atg: autophagy related; BUN: blood urea nitrogen; CCN2/CTGF: cellular communication network factor 2; CDKN2A/p16: cyclin dependent kinase inhibitor 2A; CKD: chronic kidney disease; CM: conditioned medium; COL1A1: collagen, type I, alpha 1; COL4A1: collagen, type IV, alpha 1; CQ: chloroquine; ECM: extracellular matrix; eGFR: estimated glomerular filtration rate; ELISA: enzyme-linked immunosorbent assay; FGF2: fibroblast growth factor 2; FN1: fibronectin 1; FOXO3: forkhead box O3; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HAVCR1/KIM-1: hepatitis A virus cellular receptor 1; IHC: immunohistochemistry; IRI: ischemia-reperfusion injury; ISH: in situ hybridization; LTL: lotus tetragonolobus lectin; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; PDGFB: platelet derived growth factor, B polypeptide; PPIB/cyclophilin B: peptidylprolyl isomerase B; RT-qPCR: real time-quantitative PCR; SA-GLB1/β-gal: senescence-associated galactosidase, beta 1; SASP: senescence-associated secretory phenotype; sCr: serum creatinine; SQSTM1/p62: sequestosome 1; TASCC: TOR-autophagy spatial coupling compartment; TGFB1/TGF-β1: transforming growth factor, beta 1; VIM: vimentin.
    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)是一种危及生命的疾病,是顺铂治疗最常见的副作用之一。脂肪酸氧化(FAO)是肾脏近端肾小管上皮细胞(PTEC)能量产生的主要来源,但在AKI中受到抑制。最近的工作表明,激活法尼醇X受体(FXR)可以防止AKI,但是潜在的机制仍然难以捉摸。使用顺铂诱导的AKI模型,我们发现FXR和FAO相关基因在肾脏脂质积累时显著下调。近端小管特异性或全身FXR敲除恶化,而药理激活减弱了这些作用。相反,非近端小管中的FXR敲除没有。PTEC的RNA测序表明,与顺铂治疗后的对照相比,在过表达FXR的细胞中,参与代谢途径的转录本增加。与FAO和过氧化物酶体增殖物激活受体γ(PPARγ)信号相关的特异性转录本。此外,FXR过表达或活化改善FAO并抑制顺铂处理的细胞中的细胞内脂质积累。体内研究表明,PPARγ的药理激活可以防止顺铂诱导的脂质积累,肾小管损伤和肾功能下降。然而,在顺铂治疗阶段和缺血再灌注损伤后,与对照小鼠相比,PPARγ的抑制消除了FXR的保护作用。与体内发现一致,FXR/PPARγ通过改善顺铂处理细胞中的FAO减少脂质积累。此外,肉碱棕榈酰转移酶1α的抑制作用消除了FXR在顺铂治疗小鼠中的保护作用。因此,FXR改善FAO并通过PPARγ减少肾脏PTEC中的脂质积累。因此,重建FXR/PPARγ/FAO轴可能是预防或治疗AKI的新治疗策略.
    Acute kidney injury (AKI) is a life-threatening condition that is one of most common side effects of cisplatin therapy. Fatty acid oxidation (FAO) is the main source of energy production in kidney proximal tubular epithelial cells (PTECs) but it is inhibited in AKI. Recent work demonstrated that activation of the farnesoid X receptor (FXR) protects against AKI, but the underlying mechanism remains elusive. Using a model of cisplatin-induced AKI, we found that FXR and FAO-related genes were remarkably downregulated while kidney lipid accumulated. Proximal tubule-specific or whole body FXR knockout worsened, while pharmacological activation attenuated these effects. Conversely, FXR knockout in non-proximal tubules did not. RNA-sequencing of PTECs demonstrated increased transcripts involved in metabolic pathways in cells overexpressing FXR versus control after cisplatin treatment, specifically transcripts associated with FAO and peroxisome proliferator-activated receptor-γ (PPARγ) signaling. Furthermore, FXR overexpression or activation improved FAO and inhibited intracellular lipid accumulation in cisplatin-treated cells. In vivo studies have shown that pharmacological activation of PPARγ can prevent cisplatin-induced lipid accumulation, kidney tubule injury and kidney function decline. However, inhibition of PPARγ eliminated the protective effects of FXR compared to control mice during the cisplatin treatment phase and after ischemia-reperfusion injury. Consistent with findings in vivo, FXR/PPARγ reduced lipid accumulation by improving FAO in cisplatin-treated cells. Furthermore, the inhibition of carnitine palmitoyltransferase 1α abolished the protective effect of FXR in cisplatin-treated mice. Thus, FXR improves FAO and reduced lipid accumulation via PPARγ in PTECs of the kidney. Hence, reconstruction of the FXR/PPARγ/FAO axis may be a novel therapeutic strategy for preventing or treating AKI.
    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
    与冷藏/移植相关的肾损伤是移植功能延迟和肾移植结果不良的主要因素。p53有助于缺血性和肾毒性肾损伤,但其是否参与肾脏冷藏/移植尚不清楚。这里,我们报道了肾近端小管中的p53在冷藏/移植肾损伤中起关键作用,抑制p53可以有效改善移植肾的组织学和功能。在小鼠肾脏冷藏/移植模型中,我们以冷藏时间依赖性方式检测到近端小管中p53的积累,这与肾小管损伤和细胞死亡有关。吡虫啉-α,一种药物p53抑制剂,可以减少急性肾小管损伤,细胞凋亡和炎症在冷藏/移植后24小时。从近端小管细胞中去除p53显示了类似的效果。值得注意的是,吡虫啉-α还改善了肾脏损伤,并在6天内改善了移植肾脏的功能,当时它成为受体小鼠中唯一的维持生命的肾脏。在体外,冷藏后再加温诱导培养的近端小管细胞细胞死亡,伴随着p53的激活,并被吡虫啉-α和显性阴性p53抑制。一起,这些结果支持p53在冷藏/移植肾损伤中的致病作用,并证明了p53抑制剂的治疗潜力.
    Kidney injury associated with cold storage/transplantation is a primary factor for delayed graft function and poor outcome of renal transplants. p53 contributes to both ischemic and nephrotoxic kidney injury, but its involvement in kidney cold storage/transplantation is unclear. Here, we report that p53 in kidney proximal tubules plays a critical role in cold storage/transplantation kidney injury and inhibition of p53 can effectively improve the histology and function of transplanted kidneys. In a mouse kidney cold storage/transplantation model, we detected p53 accumulation in proximal tubules in a cold storage time-dependent manner, which correlated with tubular injury and cell death. Pifithrin-α, a pharmacologic p53 inhibitor, could reduce acute tubular injury, apoptosis and inflammation at 24 h after cold storage/transplantation. Similar effects were shown by the ablation of p53 from proximal tubule cells. Notably, pifithrin-α also ameliorated kidney injury and improved the function of transplanted kidneys in 6 days when it became the sole life-supporting kidney in recipient mice. in vitro, cold storage followed by rewarming induced cell death in cultured proximal tubule cells, which was accompanied by p53 activation and suppressed by pifithrin-α and dominant-negative p53. Together, these results support a pathogenic role of p53 in cold storage/transplantation kidney injury and demonstrate the therapeutic potential of p53 inhibitors.
    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
    近端小管中脂肪酸β-氧化(FAO)的丧失是急性肾损伤和最终纤维化的关键介质。然而,FAO在近端小管损伤中的转录介质仍未得到充分研究。Krüppel类因子15(KLF15),近端小管中高度富集的锌指转录因子,马兜铃酸I(AAI)处理后,近端小管细胞显着减少,近端小管特异性损伤模型。在AAI治疗的活动期,与对照小鼠相比,Klf15的近端小管特异性敲除加剧了近端小管损伤和肾功能下降,和缺血再灌注损伤后。此外,随着近端小管损伤和肾功能下降的恶化,与对照小鼠相比,敲除小鼠在AAI治疗后的重塑阶段表现出增加的肾纤维化.肾皮质的RNA测序显示参与免疫系统和整合素信号通路的转录物增加,而包含代谢通路的转录物减少。特别是粮农组织,和PPARα信号,在AAI处理后的敲除小鼠与对照小鼠中。计算机模拟和实验性染色质免疫沉淀研究共同表明,KLF15占据了FAO关键基因的启动子区域,CPT1A和ACAA2,紧邻转录因子PPARα结合位点。虽然Klf15的损失降低了Cpt1a和Acaa2的表达,并导致粮农组织受损,KLF15的诱导部分挽救了AAI处理细胞中FAO的损失。Klf15,Ppara,Cpt1a,Acaa2的表达在其他小鼠肾损伤模型中也降低。肾小管间质KLF15与eGFR独立相关,PPARA和CPT1A在人肾活检的表达阵列中的出现。因此,Klf15的近端小管特异性丢失加剧了急性肾损伤和纤维化,可能是由于与PPARα相互作用的丧失导致FAO基因转录的丧失。
    Loss of fatty acid β-oxidation (FAO) in the proximal tubule is a critical mediator of acute kidney injury and eventual fibrosis. However, transcriptional mediators of FAO in proximal tubule injury remain understudied. Krüppel-like factor 15 (KLF15), a highly enriched zinc-finger transcription factor in the proximal tubule, was significantly reduced in proximal tubule cells after aristolochic acid I (AAI) treatment, a proximal tubule-specific injury model. Proximal tubule specific knockout of Klf15 exacerbated proximal tubule injury and kidney function decline compared to control mice during the active phase of AAI treatment, and after ischemia-reperfusion injury. Furthermore, along with worsening proximal tubule injury and kidney function decline, knockout mice exhibited increased kidney fibrosis as compared to control mice during the remodeling phase after AAI treatment. RNA-sequencing of kidney cortex demonstrated increased transcripts involved in immune system and integrin signaling pathways and decreased transcripts encompassing metabolic pathways, specifically FAO, and PPARα signaling, in knockout versus control mice after AAI treatment. In silico and experimental chromatin immunoprecipitation studies collectively demonstrated that KLF15 occupied the promoter region of key FAO genes, CPT1A and ACAA2, in close proximity to transcription factor PPARα binding sites. While the loss of Klf15 reduced the expression of Cpt1a and Acaa2 and led to compromised FAO, induction of KLF15 partially rescued loss of FAO in AAI-treated cells. Klf15, Ppara, Cpt1a, and Acaa2 expression was also decreased in other mouse kidney injury models. Tubulointerstitial KLF15 independently correlated with eGFR, PPARA and CPT1A appearance in expression arrays from human kidney biopsies. Thus, proximal tubule-specific loss of Klf15 exacerbates acute kidney injury and fibrosis, likely due to loss of interaction with PPARα leading to loss of FAO gene transcription.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号