Research progress of exosomes in diabetic nephropathy

Huang Ying, Li Ruizhao, Liang Xinling

PDF(968 KB)
Chinese Journal of Nephrology ›› 2020, Vol. 36 ›› Issue (7) : 568-572. DOI: 10.3760/cma.j.cn441217-20190925-00104
Review

Research progress of exosomes in diabetic nephropathy

Author information +
History +

Abstract

Cite this article

Download Citations
Huang Ying. , Li Ruizhao. , Liang Xinling. Research progress of exosomes in diabetic nephropathy[J]. Chinese Journal of Nephrology, 2020, 36(7): 568-572. DOI: 10.3760/cma.j.cn441217-20190925-00104.
根据国际糖尿病联盟的数据显示,2017年全球范围内糖尿病(DM)患者已达4.51亿,预计2045年将升至6.93亿[1]。糖尿病肾病(diabetic nephropathy,DN)是DM常见和严重的微血管并发症之一,是DM患者进入终末期肾脏病的主要原因,也是慢性肾脏病的主要病因[2-3]。DN早期的病理改变包括肾小球超滤和肥大,随着病变进展,出现肾小球基底膜增厚、系膜外基质积聚、足细胞减少。临床表现为肾小球滤过率降低,尿白蛋白排泄率增加,最终进展至肾小球硬化和肾衰竭。DN的发生发展与遗传、血糖升高、晚期糖基化终末产物积累、蛋白激酶C激活、氧化应激及炎性反应等因素相关[4-6],但其确切发病机制尚未完全明了。外泌体是由细胞分泌至胞外的直径约为30~150 nm的细胞外囊泡,含有细胞特异的生物活性物质。近年来,由于外泌体独特的生物学特性,外泌体成为国内外学者在探讨疾病发病机制、寻找新型分子标志物和挖掘疾病治疗新靶点的研究热点。本文就外泌体在DN的发生发展及诊疗中的研究进展进行综述。

一、 外泌体的来源和生物学特性

外泌体是细胞在生理病理状态下主动分泌的细胞外囊泡,具有维持细胞内稳态和介导细胞间信息交流的重要作用[7-8]。细胞主要通过内体模式产生外泌体,即胞内早期核内体向内出芽成熟为多泡体,多泡体与细胞质膜融合后释放外泌体。此外,外泌体还可直接经细胞质膜出芽释放或通过在胞内质膜连接隔室中出芽而延迟释放[9-10]
外泌体具有脂质双层膜结构,包裹着来源于母细胞的蛋白质、脂质、mRNA、microRNA及DNA等生物活性物质,其内容物的种类和含量随母细胞的类型和功能状态不同而发生改变[11-13]。脂质双层膜结构可以保护外泌体内的蛋白质、核酸等物质不易受外界微环境的酶降解作用。外泌体释放后,可经自分泌、旁分泌或远距分泌等途径到达靶细胞,并通过膜融合、配体受体结合或内化的方式作用于靶细胞,从而将生物活性物质转移至靶细胞或改变靶细胞的信号转导通路,最终影响细胞的功能状态[14]。外泌体具有母细胞特异性、稳定性和靶向性,广泛参与多种生理病理过程,在细胞间物质交换和信息交流中起到重要的作用。

二、 外泌体在DN发生发展中的作用

足细胞减少和肾纤维化是DN的主要病理特征。大量研究证据表明,生理病理情况下,肾细胞之间存在着丰富的物质交换与信息交流,是维持肾脏稳态或驱动疾病发生的重要原因[15-16]。外泌体作为一种细胞间通讯的新机制,病理状态下,损伤的肾细胞来源的外泌体可以通过转移mRNA、microRNA(miRNA)和转录因子等改变毗邻细胞的功能状态,在介导DN的发生与进展中起到重要作用。
1. 足细胞减少: 足细胞是高度分化的终末细胞,位于肾小球基底膜最外侧,与肾小球内皮细胞、肾小球基底膜一起构成肾小球滤过屏障,协同确保肾小球的选择性滤过功能。足细胞减少和功能障碍是DN患者出现蛋白尿的主要原因。Wang等[17]的研究表明,高糖环境下的肾小球系膜细胞可通过分泌携带转化生长因子β1(transforming growth factor-β1,TGF-β1)的外泌体激活足细胞内磷脂酰肌醇3-激酶/蛋白激酶B(PI3K/AKT)通路,诱导足细胞黏附抑制和足细胞凋亡。此外,Wu等[18]发现,高糖诱导肾小球内皮细胞分泌的外泌体使足细胞标志蛋白如nephrin、紧密连接蛋白(zonula occludens-1,ZO-1)、肾母细胞瘤蛋白1(wilms tumor 1 protein,WT 1)表达减少,而间质标志蛋白如α-平滑肌肌动蛋白(α-smooth muscle actin,α-SMA)、desmin、成纤维细胞特异蛋白1的表达增加,导致足细胞上皮-间质转化(epithelial-mesenchymal transition,EMT)和功能紊乱。
2. 肾纤维化: 肾纤维化是DN进展至慢性肾脏病和终末期肾脏病不可避免的组织病理学改变。肾纤维化的启动因素为炎性反应,表现为促炎细胞活化、炎性因子和趋化因子释放及炎细胞聚集,持续存在的慢性低度炎性反应最终导致细胞外基质沉积和肾纤维化[19]。肾小球系膜细胞和肾间质巨噬细胞在DN间质炎性反应和肾纤维化改变中扮演了重要角色。Zhu等[20]研究发现,高糖刺激下巨噬细胞分泌的外泌体可诱导肾小球系膜细胞活化与增殖,使系膜外基质α-SMA和炎性因子白细胞介素1β、肿瘤坏死因子α(TNF-α)、CC族趋化因子配体2(chemokine C-C motif ligand 2,CCL2)表达增加;动物实验中也观察到肾组织巨噬细胞浸润、系膜区基质过度沉积的病理变化。此外,高糖诱导肾小球内皮细胞分泌的外泌体也可激活肾小球系膜细胞,引起系膜外基质沉积[21]。除了高糖刺激外,持续蛋白尿也是导致DN间质炎性反应和肾纤维化的元凶之一。Lv等[22]的实验表明,暴露于白蛋白刺激的肾小管上皮细胞可通过外泌体将CCL2传递给肾间质巨噬细胞,诱导巨噬细胞活化,从而加剧肾小管间质炎性反应和肾纤维化进展。Jia等[23]的研究也发现,肾小管上皮细胞经人血清白蛋白刺激后可分泌富含miR-199a-5p的细胞外囊泡,通过Klotho/TLR4途径诱导肾间质巨噬细胞向M1表型转换,促进肾小管间质炎性反应,加速DN疾病进展。

三、 外泌体在DN诊断中的作用

目前,蛋白尿仍是临床上诊断DN和判断预后最常用的生化指标,但蛋白尿作为监测DN进展的生物标志物缺乏足够的特异性和敏感性。蛋白尿是肾小球滤过屏障受损的表现,存在于包括DN在内的许多慢性肾脏病中[24]。此外,有研究发现,DN患者的肾脏结构损害可先于微量白蛋白尿出现[25],且部分患者虽维持在微量白蛋白尿水平,但其肾功能已进行性下降[26]。这些证据表明蛋白尿不能完全正确地反映DN的肾脏损害程度。因此,探索更具特异性与敏感性的分子标志物对DN的早期诊断和病情监测有重要意义。尿外泌体包含的蛋白质、核酸等物质,可以反映母细胞的功能状态。从尿液中提取外泌体并分析其成分变化已经成为DN分子标志物研究的一个新方向。
1. 足细胞损伤相关分子标志物: WT 1是足细胞标志性蛋白。Kalani等[27]发现早期DN患者尿外泌体中WT 1的表达水平随患者肾功能下降而升高,提示WT 1可作为DN早期肾功能的监测指标。此外,尿外泌体中WT 1 mRNA水平可以预测DN患者估算肾小球滤过率(eGFR)的下降速率[28]
2. 肾间质炎性反应或肾纤维化相关分子标志物: Lv等[29]研究发现,在急性肾损伤和慢性肾脏病疾病模型中,肾小管上皮细胞来源的外泌体内miR-19b-3p与肾小管间质炎性反应有关。随后,Lv等[29]对28例经肾活检证实DN患者和15例Ⅱ型糖尿病患者的尿标本对比研究发现,DN患者尿外泌体内miR-19b-3p表达显著增高,并且与DN患者肾间质炎性反应严重程度呈正相关关系,提示miR-19b-3p可作为监测DN的分子标志物。除此之外,miR-200b[30]、miR-29c[31]、CD2相关蛋白[32]也可作为肾纤维化的潜在分子标志物。
3. 参与DN发病机制的信号通路相关分子标志物: Xie等[33]研究发现,与Ⅱ型糖尿病患者比较,DN患者尿外泌体内miR-362-3p、miR-877-3p和miR-150-5p表达上调,miR-15a-5p表达下调。这些变化的miRNA可能通过p53、mTOR和AMPK信号通路参与调控DN,是早期DN潜在的生物标志物。还有研究发现,DN 患者尿外泌体中miR-145 和 miR-320c 表达上调,这些miRNA与调控TGF-β1介导的信号通路有关,可能也是DN潜在的分子标志物[34-35]
4. 其他分子标志物: 尿外泌体中的let-7c-5p、水通道蛋白(water channel aquaporin,AQP)5和AQP2、亮氨酸氨基肽酶和二肽基肽酶Ⅳ以及钙调素等也被认为可能是DN潜在的分子标志物[36-39]。尿外泌体能动态且稳定地携带来源于母细胞的蛋白质、核酸等生物信息,不仅可作为诊断和监测DN的分子标志物,还有助于进一步探索DN发病的分子机制。因此,尿外泌体在挖掘DN分子标志物方面有着独特的优势和研究前景。

四、 外泌体在DN治疗中的作用

随着干细胞再生医学的发展,干细胞来源的外泌体在治疗方面的DN潜力也受到了广泛关注。干细胞分泌的外泌体含有母细胞来源的生长因子、细胞因子和miRNA等生物活性物质,因此具有类似干细胞的免疫调节、抗细胞凋亡和损伤修复等特性[40]。Jin等[41]研究发现,脂肪来源干细胞分泌的外泌体可以减少DN小鼠蛋白尿、血肌酐和血尿素氮水平,减轻小鼠肾脏病理损伤,且在体外实验中可以逆转高糖诱导的足细胞凋亡。Jin等[41]的研究表明,脂肪来源干细胞分泌的外泌体可通过抑制足细胞内mTOR信号通路诱导自噬流增加,从而减少足细胞凋亡。人尿液来源干细胞、间充质干细胞(mesenchymal stem cell,MSC)分泌的外泌体也具有减少足细胞凋亡的功能效应[42-43]。Nagaishi等[44]用MSC干预DN小鼠,发现MSC可抑制小鼠TNF-α和TGF-β1表达,减少ZO-1降解,从而抑制肾小管上皮细胞EMT和肾纤维化,表明该作用与MSC的外泌体旁分泌途径有关。综上所述,干细胞来源的外泌体可通过抗细胞凋亡、抗炎和抗纤维化等多种效应改善DN的肾功能和肾脏病理损伤。
干细胞外泌体具有天然的治疗疾病作用,也是理想的药物治疗载体。外泌体具有良好的靶向特性和生物相容性,可通过共培养、转染和电穿孔等方式,将具有治疗作用的分子或药物装载进入外泌体并使之靶向损伤部位,从而产生治疗效应[45]。近年来,通过构建药物载体外泌体用于肿瘤、心血管疾病和神经退行性疾病等疾病的治疗方面的研究日益增多,显现出广阔的应用前景[46]。Tang等[47]的研究显示,装载了地塞米松的细胞外囊泡不仅可有效靶向有炎性损伤的肾脏,且与单纯的地塞米松治疗相比,装载了地塞米松的细胞外囊泡显示出更强的抗炎和抗纤维化作用。相信随着研究不断深入,药物载体外泌体也将有望成为DN治疗的新型治疗手段。

五、 总结与展望

DN是DM常见而严重的微血管并发症,是DM患者进入终末期肾脏病的主要原因,但其发病机制复杂,至今未能完全明确,临床上也尚缺乏理想有效的诊疗方法。现有的研究表明,外泌体在DN的发生发展、诊断与治疗中扮演重要角色。外泌体携带着丰富的生物分子,可与邻近或远距离的靶细胞作用,是介导细胞间信息交流的天然媒介,这为我们探讨在DN发病过程中肾脏与其他系统的对话交流、肾脏固有细胞之间的相互作用提供了新思路。对外泌体的转录组学和蛋白质组学分析,也有助于我们发现更多潜在的致病分子及其致病相关机制。此外,阻断含有致病分子的外泌体释放、使用干细胞来源外泌体、构建药物载体外泌体等手段,也将成为学者们探索有效防治DN的研究重点。随着理论知识的逐渐丰富和研究的逐渐深入,外泌体在糖尿病肾病发生发展中所起的作用将不断被揭示,外泌体也有望成为早期诊断和有效防治DN极具价值的新靶点。

References

[1]
Cho NH, Shaw JE, Karuranga S, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045[J]. Diabetes Res Clin Pract, 2018, 138: 271-281. DOI: 10.1016/j.diabres.2018.02.023.
[2]
Tuttle KR, Bakris GL, Bilous RW, et al. Diabetic kidney disease: a report from an ADA consensus conference[J]. Diabetes Care, 2014, 37(10): 2864-2883. DOI: 10.2337/dc14-1296.
The incidence and prevalence of diabetes mellitus have grown significantly throughout the world, due primarily to the increase in type 2 diabetes. This overall increase in the number of people with diabetes has had a major impact on development of diabetic kidney disease (DKD), one of the most frequent complications of both types of diabetes. DKD is the leading cause of end-stage renal disease (ESRD), accounting for approximately 50% of cases in the developed world. Although incidence rates for ESRD attributable to DKD have recently stabilized, these rates continue to rise in high-risk groups such as middle-aged African Americans, Native Americans, and Hispanics. The costs of care for people with DKD are extraordinarily high. In the Medicare population alone, DKD-related expenditures among this mostly older group were nearly $25 billion in 2011. Due to the high human and societal costs, the Consensus Conference on Chronic Kidney Disease and Diabetes was convened by the American Diabetes Association in collaboration with the American Society of Nephrology and the National Kidney Foundation to appraise issues regarding patient management, highlighting current practices and new directions. Major topic areas in DKD included 1) identification and monitoring, 2) cardiovascular disease and management of dyslipidemia, 3) hypertension and use of renin-angiotensin-aldosterone system blockade and mineralocorticoid receptor blockade, 4) glycemia measurement, hypoglycemia, and drug therapies, 5) nutrition and general care in advanced-stage chronic kidney disease, 6) children and adolescents, and 7) multidisciplinary approaches and medical home models for health care delivery. This current state summary and research recommendations are designed to guide advances in care and the generation of new knowledge that will meaningfully improve life for people with DKD. © 2014 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.
[3]
Zhang L, Long J, Jiang W, et al. Trends in chronic kidney disease in China[J]. N Engl J Med, 2016, 375(9): 905-906. DOI: 10.1056/NEJMc1602469.
[4]
Carpena MP, Rados DV, Sortica DA, et al. Genetics of diabetic nephropathy[J]. Arq Bras Endocrinol Metabol, 2010, 54(3): 253-261. DOI: 10.1590/s0004-27302010000300002.
The increasing prevalence of diabetes mellitus has led to a growing number of chronic complications including diabetic nephropathy (DN). In addition to its high prevalence, DN is associated with high morbidity and mortality especially due to cardiovascular diseases. It is well established that genetic factors play a role in the pathogenesis of DN and genetically susceptible individuals can develop it after being exposed to environmental factors. DN is probably a complex, polygenic disease. Two main strategies have been used to identify genes associated to DN: analysis of candidate genes, and more recently genome-wide scan. Great efforts have been made to identify these main genes, but results are still inconsistent with different genes associated to a small effect in specific populations. The identification of the main genes would allow the detection of those individuals at high risk for DN and better understanding of its pathophysiology as well.
[5]
Kanwar YS, Sun L, Xie P, et al. A glimpse of various pathogenetic mechanisms of diabetic nephropathy[J]. Annu Rev Pathol, 2011, 6: 395-423. DOI: 10.1146/annurev.pathol.4.110807.092150.
Diabetic nephropathy is a well-known complication of diabetes and is a leading cause of chronic renal failure in the Western world. It is characterized by the accumulation of extracellular matrix in the glomerular and tubulointerstitial compartments and by the thickening and hyalinization of intrarenal vasculature. The various cellular events and signaling pathways activated during diabetic nephropathy may be similar in different cell types. Such cellular events include excessive channeling of glucose intermediaries into various metabolic pathways with generation of advanced glycation products, activation of protein kinase C, increased expression of transforming growth factor β and GTP-binding proteins, and generation of reactive oxygen species. In addition to these metabolic and biochemical derangements, changes in the intraglomerular hemodynamics, modulated in part by local activation of the renin-angiotensin system, compound the hyperglycemia-induced injury. Events involving various intersecting pathways occur in most cell types of the kidney.
[6]
Navarro-González JF, Mora-Fernández C. The role of inflammatory cytokines in diabetic nephropathy[J]. J Am Soc Nephrol, 2008, 19(3): 433-442. DOI: 10.1681/ASN.2007091048.
Cytokines act as pleiotropic polypeptides regulating inflammatory and immune responses through actions on cells. They provide important signals in the pathophysiology of a range of diseases, including diabetes mellitus. Chronic low-grade inflammation and activation of the innate immune system are closely involved in the pathogenesis of diabetes and its microvascular complications. Inflammatory cytokines, mainly IL-1, IL-6, and IL-18, as well as TNF-alpha, are involved in the development and progression of diabetic nephropathy. In this context, cytokine genetics is of special interest to combinatorial polymorphisms among cytokine genes, their functional variations, and general susceptibility to diabetic nephropathy. Finally, the recognition of these molecules as significant pathogenic mediators in diabetic nephropathy leaves open the possibility of new potential therapeutic targets.
[7]
Camussi G, Deregibus MC, Bruno S, et al. Exosomes/microvesicles as a mechanism of cell-to-cell communication[J]. Kidney Int, 2010, 78(9): 838-848. DOI: 10.1038/ki.2010.278.
Microvesicles (MVs) are circular fragments of membrane released from the endosomal compartment as exosomes or shed from the surface membranes of most cell types. An increasing body of evidence indicates that they play a pivotal role in cell-to-cell communication. Indeed, they may directly stimulate target cells by receptor-mediated interactions or may transfer from the cell of origin to various bioactive molecules including membrane receptors, proteins, mRNAs, microRNAs, and organelles. In this review we discuss the pleiotropic biologic effects of MVs that are relevant for communication among cells in physiological and pathological conditions. In particular, we discuss their potential involvement in inflammation, renal disease, and tumor progression, and the evidence supporting a bidirectional exchange of genetic information between stem and injured cells. The transfer of gene products from injured cells may explain stem cell functional and phenotypic changes without the need of transdifferentiation into tissue cells. On the other hand, transfer of gene products from stem cells may reprogram injured cells to repair damaged tissues.
[8]
Baixauli F, López-Otín C, Mittelbrunn M. Exosomes and autophagy: coordinated mechanisms for the maintenance of cellular fitness[J]. Front Immunol, 2014, 5: 403. DOI: 10.3389/fimmu.2014.00403.
Conditions resulting from loss of cellular homeostasis, including oxidative stress, inflammation, protein aggregation, endoplasmic reticulum stress, metabolic stress, and perturbation of mitochondrial function, are common to many pathological disorders and contribute to aging. Cells face these stress situations by engaging quality control mechanisms aimed to restore cellular homeostasis and preserve cell viability. Among them, the autophagylysosomal pathway mediates the specific degradation of damaged proteins and organelles, and its proper function is related to cellular protection and increased life span in many model organisms. Besides autophagy, increasing evidence underscores a role for exosomes in the selective secretion of harmful/damaged proteins and RNAs and thus in the maintenance of cellular fitness. In this perspective article, we discuss the emerging function of exosomes as a means of alleviating intracellular stress conditions, and how secretion of harmful or unwanted material in exosomes, in coordination with the autophagy-lysosomal pathway, is essential to preserve intracellular protein and RNA homeostasis. Finally, we provide an overview about the consequences of the spreading of the exosome content in physiological and pathological situations, and suggest putative therapeutic strategies for these exosome-mediated alterations.
[9]
Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles[J]. Annu Rev Cell Dev Biol, 2014, 30: 255-289. DOI: 10.1146/annurev-cellbio-101512-122326.
In the 1980s, exosomes were described as vesicles of endosomal origin secreted from reticulocytes. Interest increased around these extracellular vesicles, as they appeared to participate in several cellular processes. Exosomes bear proteins, lipids, and RNAs, mediating intercellular communication between different cell types in the body, and thus affecting normal and pathological conditions. Only recently, scientists acknowledged the difficulty of separating exosomes from other types of extracellular vesicles, which precludes a clear attribution of a particular function to the different types of secreted vesicles. To shed light into this complex but expanding field of science, this review focuses on the definition of exosomes and other secreted extracellular vesicles. Their biogenesis, their secretion, and their subsequent fate are discussed, as their functions rely on these important processes.
[10]
Pegtel DM, Gould SJ. Exosomes[J]. Annu Rev Biochem, 2019, 88: 487-514. DOI: 10.1146/annurev-biochem-013118-111902.
Exosomes are small, single-membrane, secreted organelles of ∼30 to ∼200 nm in diameter that have the same topology as the cell and are enriched in selected proteins, lipids, nucleic acids, and glycoconjugates. Exosomes contain an array of membrane-associated, high-order oligomeric protein complexes, display pronounced molecular heterogeneity, and are created by budding at both plasma and endosome membranes. Exosome biogenesis is a mechanism of protein quality control, and once released, exosomes have activities as diverse as remodeling the extracellular matrix and transmitting signals and molecules to other cells. This pathway of intercellular vesicle traffic plays important roles in many aspects of human health and disease, including development, immunity, tissue homeostasis, cancer, and neurodegenerative diseases. In addition, viruses co-opt exosome biogenesis pathways both for assembling infectious particles and for establishing host permissiveness. On the basis of these and other properties, exosomes are being developed as therapeutic agents in multiple disease models.
[11]
Zhang W, Zhou X, Zhang H, et al. Extracellular vesicles in diagnosis and therapy of kidney diseases[J]. Am J Physiol Renal Physiol, 2016, 311(5): F844-F851. DOI: 10.1152/ajprenal.00429.2016.
Extracellular vesicles (EV) are endogenously produced, membrane-bound vesicles that contain various molecules. Depending on their size and origins, EVs are classified into apoptotic bodies, microvesicles, and exosomes. A fundamental function of EVs is to mediate intercellular communication. In kidneys, recent research has begun to suggest a role of EVs, especially exosomes, in cell-cell communication by transferring proteins, mRNAs, and microRNAs to recipient cells as nanovectors. EVs may mediate the cross talk between various cell types within kidneys for the maintenance of tissue homeostasis. They may also mediate the cross talk between kidneys and other organs under physiological and pathological conditions. EVs have been implicated in the pathogenesis of both acute kidney injury and chronic kidney diseases, including renal fibrosis, end-stage renal disease, glomerular diseases, and diabetic nephropathy. The release of EVs with specific molecular contents into urine and plasma may be useful biomarkers for kidney disease. In addition, EVs produced by cultured cells may have therapeutic effects for these diseases. However, the role of EVs in kidney diseases is largely unclear, and the mechanism underlying EV production and secretion remains elusive. In this review, we introduce the basics of EVs and then analyze the present information about the involvement, diagnostic value, and therapeutic potential of EVs in major kidney diseases.
[12]
Parolini I, Federici C, Raggi C, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells[J]. J Biol Chem, 2009, 284(49): 34211-34222. DOI: 10.1074/jbc.M109.041152.
Exosomes secreted by normal and cancer cells carry and deliver a variety of molecules. To date, mechanisms referring to tumor exosome trafficking, including release and cell-cell transmission, have not been described. To gain insight into this, exosomes purified from metastatic melanoma cell medium were labeled with a lipid fluorescent probe, R18, and analyzed by spectrofluorometry and confocal microscopy. A low pH condition is a hallmark of tumor malignancy, potentially influencing exosome release and uptake by cancer cells. Using different pH conditions as a modifier of exosome traffic, we showed (i) an increased exosome release and uptake at low pH when compared with a buffered condition and (ii) exosome uptake by melanoma cells occurred by fusion. Membrane biophysical analysis, such as fluidity and lipid composition, indicated a high rigidity and sphingomyelin/ganglioside GM3 (N-acetylneuraminylgalactosylglucosylceramide) content in exosomes released at low pH. This was likely responsible for the increased fusion efficiency. Consistent with these results, pretreatment with proton pump inhibitors led to an inhibition of exosome uptake by melanoma cells. Fusion efficiency of tumor exosomes resulted in being higher in cells of metastatic origin than in those derived from primary tumors or normal cells. Furthermore, we found that caveolin-1, a protein involved in melanoma progression, is highly delivered through exosomes released in an acidic condition. The results of our study provide the evidence that exosomes may be used as a delivery system for paracrine diffusion of tumor malignancy, in turn supporting the importance of both exosomes and tumor pH as key targets for future anti-cancer strategies.
[13]
郭艳霞, 宋秀珍, 周秋根. 缺氧对肾小管上皮细胞分泌外泌体的影响[J]. 中华肾脏病杂志, 2015, 31(12): 911-918. DOI: 10.3760/cma.j.issn.1001-7097.2015.12.006.
目的&nbsp;&nbsp;&nbsp; 观察缺氧对肾小管上皮细胞分泌外泌体的影响,探讨外泌体在缺氧致肾脏损伤中的作用及机制。 方法&nbsp;&nbsp;&nbsp; (1)常氧(21% O<sub>2</sub>)及缺氧(1% O<sub>2</sub>)分别处理大鼠肾小管上皮细胞(NRK-52E)48 h,收集细胞上清液并使用高速梯度离心法分离外泌体。采用透射电镜、纳米示踪分析、Western印迹、蛋白浓度定量鉴定并比较两组外泌体的基本特性。(2)在共培养实验中,以不同浓度(1、10、50、100、300 mg/L)的常氧外泌体、缺氧外泌体分别干预脂多糖(LPS)诱导的大鼠原代腹腔巨噬细胞,使用实时荧光定量PCR与酶联免疫吸附试验(ELISA)法分别检测巨噬细胞白细胞介素6(IL-6)、肿瘤坏死因子&alpha;(TNF-&alpha;)、诱导型氮氧化物合酶(iNOS)水平;使用Western印迹法检测巨噬细胞磷酸化(p)STAT/STAT及细胞因子信号传导抑制蛋白1(SOCS1)的蛋白表达;最后,使用实时荧光定量PCR法检测常氧外泌体与缺氧外泌体中炎性反应相关微RNA(microRNA,miR)的表达差异。 结果&nbsp;&nbsp;&nbsp; (1)离心得到的囊泡具有外泌体典型的结构,粒径小于150 nm,表达外泌体标志蛋白CD63,说明分离得到外泌体。缺氧对肾小管上皮细胞分泌的外泌体形态、粒径分布比例无明显影响,但提高了外泌体的分泌量。(2)缺氧外泌体相比于常氧外泌体促进了LPS诱导的M1型巨噬细胞IL-6、TNF-&alpha;、iNOS 的表达和分泌(均P<0.01),同时提高STAT的磷酸化水平并减少SOCS1的蛋白表达(均P<0.01);对炎性反应相关microRNA检测发现缺氧外泌体中miR-155、miR-27a表达量较常氧外泌体明显升高(P<0.05)。 结论&nbsp;&nbsp;&nbsp; 缺氧可改变外泌体的生物学功能,表现为协同促进LPS诱导的M1型巨噬细胞的表型转化,这可能是慢性肾脏病微炎性反应状态持续的原因之一。
[14]
Boriachek K, Islam MN, Moller A, et al. Biological functions and current advances in isolation and detection strategies for exosome nanovesicles[J]. Small, 2018, 14(6): 10. DOI: 1002/smll.201702153.
[15]
Lennon R, Hosawi S. Glomerular cell crosstalk[J]. Curr Opin Nephrol Hypertens, 2016, 25(3): 187-193. DOI: 10.1097/MNH.0000000000000221.
[16]
Wang J, Zhong J, Yang HC, et al. Cross talk from tubules to glomeruli[J]. Toxicol Pathol, 2018, 46(8): 944-948. DOI: 10.1177/0192623318796784.
Tubular injury sensitizes glomeruli to injury. We review potential mechanisms of this tubuloglomerular cross talk. In the same nephron, tubular injury can cause stenosis of the glomerulotubular junction and finally result in atubular glomeruli. Tubular injury also affects glomerular filtration function through tubuloglomerular feedback. Progenitor cells, that is, parietal epithelial cells and renin positive cells, can be involved in repair of injured glomeruli and also may be modulated by tubular injury. Loss of nephrons induces additional workload and stress on remaining nephrons. Hypoxia and activation of the renin-angiotensin-aldosterone system induced by tubular injury also modulate tubuloglomerular cross talk. Therefore, effective therapies in chronic kidney disease may need to aim to interrupt this deleterious tubuloglomerular cross talk.
[17]
Wang YY, Tang LQ, Wei W. Berberine attenuates podocytes injury caused by exosomes derived from high glucose-induced mesangial cells through TGFβ1-PI3K/AKT pathway[J]. Eur J Pharmacol, 2018, 824: 185-192. DOI: 10.1016/j.ejphar.2018.01.034.
[18]
Wu X, Gao Y, Xu L, et al. Exosomes from high glucose-treated glomerular endothelial cells trigger the epithelial-mesenchymal transition and dysfunction of podocytes[J]. Sci Rep, 2017, 7(1): 9371. DOI: 10.1038/s41598-017-09907-6.
New data indicate that abnormal glomerular endothelial cell (GEC)-podocyte crosstalk plays a critical role in diabetic nephropathy (DN). The aim of our study is to investigate the role of exosomes from high glucose (HG)-treated GECs in the epithelial-mesenchymal transition (EMT) and dysfunction of podocytes. In this study, exosomes were extracted from GEC culture supernatants and podocytes were incubated with the GEC-derived exosomes. Here, we demonstrate that HG induces the endothelialmesenchymal transition (EndoMT) of GECs and HG-treated cells undergoing the EndoMT secrete more exosomes than normal glucose (NG)-treated GECs. We show that GEC-derived exosomes can be internalized by podocytes and exosomes from HG-treated cells undergoing an EndoMT-like process can trigger the podocyte EMT and barrier dysfunction. Our study reveals that TGF-beta 1 mRNA is enriched in exosomes from HG-treated GECs and probably mediates the EMT and dysfunction of podocytes. In addition, our experimental results illustrate that canonical Wnt/beta-catenin signaling is involved in the exosome-induced podocyte EMT. Our findings suggest the importance of paracrine communication via exosomes between cells undergoing the EndoMT and podocytes for renal fibrosis in DN. Thus, protecting GECs from the EndoMT and inhibiting TGF-beta 1-containing exosomes release from GECs is necessary to manage renal fibrosis in DN.
[19]
Lv W, Booz GW, Wang Y, et al. Inflammation and renal fibrosis: recent developments on key signaling molecules as potential therapeutic targets[J]. Eur J Pharmacol, 2018, 820: 65-76. DOI: 10.1016/j.ejphar.2017.12.016.
Chronic kidney disease (CKD) is a major public health issue. At the histological level, renal fibrosis is the final common pathway of progressive kidney disease irrespective of the initial injury. Considerable evidence now indicates that renal inflammation plays a central role in the initiation and progression of CKD. Some of the inflammatory signaling molecules involved in CKD include: monocyte chemoattractant protein-1 (MCP-1), bradykinin B receptor (BR), nuclear factor κB (NF-κB), tumor necrosis factor-α (TNFα), transforming growth factor β (TGF-β), and platelet-derived growth factor (PDGF). Multiple antifibrotic factors, such as interleukin-10 (IL-10), interferon-γ (IFN-γ), bone morphogenetic protein-7 (BMP-7), hepatocyte growth factor (HGF) are also downregulated in CKD. Therefore, restoration of the proper balance between pro- and antifibrotic signaling pathways could serve as a guiding principle for the design of new antifibrotic strategies that simultaneously target many pathways. The purpose of this review is to summarize the existing body of knowledge regarding activation of cytokine pathways and infiltration of inflammatory cells as a starting point for developing novel antifibrotic therapies to prevent progression of CKD.Copyright © 2017 Elsevier B.V. All rights reserved.
[20]
Zhu QJ, Zhu M, Xu XX, et al. Exosomes from high glucose-treated macrophages activate glomerular mesangial cells via TGF-beta1/Smad3 pathway in vivo and in vitro[J]. FASEB J, 2019, 33(8): 9279-9290. DOI: 10.1096/fj.201802427RRR.
[21]
Wu XM, Gao YB, Cui FQ, et al. Exosomes from high glucose-treated glomerular endothelial cells activate mesangial cells to promote renal fibrosis[J]. Biology Open, 2016, 5(4): 484-491. DOI: 10.1242/bio.015990.
The interaction between glomerular endothelial cells (GECs) and glomerular mesangial cells (GMCs) is an essential aspect of diabetic nephropathy (DN). Therefore, understanding how GECs communicate with GMCs in the diabetic environment is crucial for the development of new targets for the prevention and treatment of DN. Exosomes, nanometer-sized extracellular membrane vesicles secreted by various cell types, play important roles in cell-to-cell communication via the transfer of mRNA, microRNA and protein. In this study, we demonstrate that high glucose (HG)-treated GECs secrete a higher number of exosomes highly enriched in TGF-β1 mRNA compared with normal glucose (NG)-treated GECs. Exosomes released by HG-treated GECs can promote α-smooth muscle actin (α-SMA) expression, proliferation and extracellular matrix protein overproduction in GMCs through the TGF-β1/Smad3 signaling pathway. Thus, we provide new insights into the pathogenesis of DN that involves intercellular transfer of TGF-β1 mRNA in the GEC-to-GMC direction via exosomes.
[22]
Lv LL, Feng Y, Wen Y, et al. Exosomal CCL2 from tubular epithelial cells is critical for albumin-induced tubulointerstitial inflammation[J]. J Am Soc Nephrol, 2018, 29(3): 919-935. DOI: 10.1681/ASN.2017050523.
Albuminuria is a key instigator of tubulointerstitial inflammation associated with CKD, but the mechanism through which filtered albumin propagates renal injury remains unclear. In this study, we explored the role in this process of exosome mRNA released from tubular epithelial cells (TECs). Compared with control mice, acute and chronic kidney injury models had more exosomes containing inflammatory cytokine mRNA, particularly the chemokine CCL2, in kidneys and urine. In vitro stimulation of TECs with BSA recapitulated this finding. Notably, the internalization of purified TEC exosomes by cultured macrophages increased if TECs were exposed to BSA. Macrophage internalization of exosomes from BSA-treated TECs led to an enhanced inflammatory response and macrophage migration, but CCL2 silencing in TECs prevented these effects. Using a GFP-CCL2 fusion mRNA construct, we observed direct transfer of CCL2 mRNA from TEC exosomes to macrophages. Mice subjected to tail vein injection of purified BSA-treated TEC exosomes developed tubular injury with renal inflammatory cell infiltration. However, injection of exosomes from BSA-treated CCL2-deficient TECs induced less severe kidney inflammation. Finally, in patients with IgA nephropathy, the increase of proteinuria correlated with augmented urinary excretion of exosomes with exaggerated expression of CCL2 mRNA. Moreover, the level of CCL2 mRNA in urinary exosomes correlated closely with levels of renal interstitial macrophage infiltration in these patients. Our studies demonstrate that the increasing release of exosomes that transfer CCL2 mRNA from TECs to macrophages constitutes a critical mechanism of albumin-induced tubulointerstitial inflammation.
[23]
Jia YJ, Zheng ZJ, Xue M, et al. Extracellular vesicles from albumin-induced tubular epithelial cells promote the M1 macrophage phenotype by targeting klotho[J]. Mol Ther, 2019, 27(8): 1452-1466. DOI: 10.1016/j.ymthe.2019.05.019.
Albumin absorbed by renal tubular epithelial cells induces inflammation and plays a key role in promoting diabetic kidney disease (DKD) progression. Macrophages are prominent inflammatory cells in the kidney, and their role there is dependent on their phenotypes. However, whether albuminuria influences macrophage phenotypes and underlying mechanisms during the development of DKD is still unclear. We found that M1 macrophage-related markers were increased in diabetes mellitus (DM) mouse renal tissues with the development of DKD, and coculture of extracellular vesicles (EVs) from human serum albumin (HSA)-induced HK-2 cells with macrophages induced macrophage M1 polarization in the presence of lipopolysaccharide (LPS). Through a bioinformatic analysis, miR-199a-5p was selected and found to be increased in EVs from HSA-induced HK-2 cells and in urinary EVs from DM patients with macroalbuminuria. Tail-vein injection of DM mice with EVs from HSA-induced HK-2 cells induced kidney macrophage M1 polarization and accelerated the progression of DKD through miR-199a-5p. miR-199a-5p exerted its effect by targeting Klotho, and Klotho induced macrophage M2 polarization through the Toll-like receptor 4 (TLR4) pathway both in vivo and in vitro. In summary, miR-199a-5p from HSA-stimulated HK-2 cell-derived EVs induces M1 polarization by targeting the Klotho/TLR4 pathway and further accelerates the progression of DKD.Copyright © 2019 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.
[24]
Glassock RJ. Is the presence of microalbuminuria a relevant marker of kidney disease?[J]. Curr Hypertens Rep, 2010, 12(5): 364-368. DOI: 10.1007/s11906-010-0133-3.
Levels of urinary albumin excretion that are below the usual limit of detection by qualitative testing, but are above normal levels (microalbuminuria; MA), can be readily identified by simple measures, such as the urinary albumin to creatinine ratio in untimed urine samples. Such measurements, particularly when combined with assessment of estimated glomerular filtration rate (eGFR), have utility as biomarkers for enhanced risk of all-cause mortality, cardiovascular events, progressive chronic kidney disease, and end-stage renal disease in diabetic and nondiabetic subjects. However, it is controversial whether "isolated" MA (MA in the absence of a clear reduction in eGFR, urine sediment abnormalities, or structural renal disease) should be regarded as kidney disease. Such MA could also be regarded as a manifestation of a diffuse endothelial (microvascular) injury and thereby collateral kidney damage. This article reviews the current evidence concerning MA as a marker of kidney disease or kidney damage.
[25]
Chavers BM, Bilous RW, Ellis EN, et al. Glomerular lesions and urinary albumin excretion in type I diabetes without overt proteinuria[J]. N Engl J Med, 1989, 320(15): 966-970. DOI: 10.1056/NEJM198904133201503.
[26]
Perkins BA, Ficociello LH, Roshan B, et al. In patients with type 1 diabetes and new-onset microalbuminuria the development of advanced chronic kidney disease may not require progression to proteinuria[J]. Kidney Int, 2010, 77(1): 57-64. DOI: 10.1038/ki.2009.399.
We sought to study new-onset microalbuminuria, its progression, and the decline of renal function in patients with type 1 diabetes. Using a cohort of 109 patients who developed new-onset microalbuminuria in the first 4 years following enrollment in the 1st Joslin Kidney Study, we simultaneously tracked the change in their renal function and urinary albumin excretion. Of these, 79 patients were followed for an average of 12 years after microalbuminuria onset, wherein their glomerular filtration rate was estimated by the Modification of Diet in Renal Disease Study formula and compared with their microalbuminuria and proteinuria. The concordance between these outcomes was weak. Only 12 of the 23 patients who progressed to advanced (stage 3-5) chronic kidney disease developed proteinuria, which, in general, did not precede but accompanied the progression to advanced chronic kidney disease. The remaining 11 patients who developed advanced disease had persistent microalbuminuria or returned to normal albuminuria. Thus, we found that one-third of patients with type 1 diabetes developed advanced chronic kidney disease relatively soon after the onset of microalbuminuria and this was not conditional on the presence of proteinuria. Contrary to the existing concept of early nephropathy in type 1 diabetes, less emphasis should be placed on the mechanisms of progression to proteinuria and more placed on mechanisms initiating and promoting the early decline of renal function that eventually progresses to advanced chronic kidney disease.
[27]
Kalani A, Mohan A, Godbole MM, et al. Wilm's tumor-1 protein levels in urinary exosomes from diabetic patients with or without proteinuria[J]. PLoS One, 2013, 8(3): e60177. DOI: 10.1371/journal.pone.0060177.
[28]
Abe H, Sakurai A, Ono H, et al. Urinary exosomal mRNA of WT1 as diagnostic and prognostic biomarker for diabetic nephropathy[J]. J Med Invest, 2018, 65(3-4): 208-215. DOI: 10.2152/jmi.65.208.
[29]
Lv LL, Feng Y, Wu M, et al. Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury[J]. Cell Death Differ, 2019, 27(1): 210-226. DOI: 10.1038/s41418-019-0349-y.
[30]
Yu Y, Bai F, Qin N, et al. Non-proximal renal tubule-derived urinary exosomal miR-200b as a biomarker of renal fibrosis[J]. Nephron, 2018, 139(3): 269-282. DOI: 10.1159/000487104.
&lt;b&gt;<i>Background:</i>&lt;/b&gt; Renal fibrosis is a common outcome of nearly all kinds of chronic kidney disease (CKD) and eventually progresses to end-stage renal disease. The identification of an optimal biomarker of renal fibrosis to replace the invasive renal biopsy will have important clinical implications. &lt;b&gt;<i>Methods:</i>&lt;/b&gt; We isolated urinary exosomes from 50 participants and examined the exosomal protein content and particle number in 38 CKD patients with different degrees of renal fibrosis and in 12 normal individuals. We examined the levels of exosomal microRNAs (miRNAs), namely, miR-200a, miR-200b, miR-200c, miR-141, miR-429, miR-29a, miR-29b, miR-29c, miR-192, and miR-21, by sorting the exosomes and comparing the levels of proximal tubular, non-proximal tubular, and total exosomal miR-200b. &lt;b&gt;<i>Results:</i>&lt;/b&gt; The exosome content was higher in the CKD group, but no differences were evident among the mild, moderate, and severe fibrosis groups. Among the 10 exosomal miRNAs, miR-200b was lower in the CKD group than in the normal group and decreased more significantly with fibrosis progression as well as in IgA nephropathy and diabetic kidney disease. CD13<sup>+</sup> CD63<sup>+</sup> exosomes constituted 18.6% of all urinary exosomes. Sorting the proximal tubular exosomes with the CD13 protein marker revealed that miR-200b in the CD13<sup>+</sup> group was extremely low; however, the result was significantly different in the CD13<sup>–</sup> group but not in the CD13<sup>+</sup> group. The magnitude of the decline was greater in the CD13<sup>–</sup> groups than in the non-sorted whole groups between the fibrosis and normal patients. &lt;b&gt;<i>Conclusions:</i>&lt;/b&gt; Non-proximal renal tubule-derived urinary exosomal miR-200b is a biomarker of renal fibrosis. Exosomes can be used as a liquid biopsy and may replace the traditional invasive renal biopsy in the diagnosis of renal fibrosis.
[31]
Lv LL, Cao YH, Ni HF, et al. microRNA-29c in urinary exosome/microvesicle as a biomarker of renal fibrosis[J]. Am J Physiol Renal Physiol, 2013, 305(8): F1220-F1227. DOI: 10.1152/ajprenal.00148.2013.
Micro (mi)RNAs are frequently dysregulated in the development of renal fibrosis. Exosomes are small membrane vesicles that could be isolated from urine secreted from all nephron segments. Here we sought to observe for the first time whether miRNA in urine exosome could serve as a potential biomarker of renal fibrosis. Urine samples were collected from 32 chronic kidney disease (CKD) patients who underwent kidney biopsy and 7 controls. Exosome was isolated and confirmed by immunogold staining of exosome marker. Members of miR-29, miR-200, and RNU6B as endogenous control were detected by RT quantitative PCR. Electronic microscopy verified a typical shape of exosome with average size of 65.1 nm and labeled it with anti-CD9 and anti-aquaporin 2 antibody. Members of miR-29 and miR-200 are readily measured with reduced levels compared with controls ( P &lt; 0.05) and can robustly distinguish CKD from controls [area under the curve (AUC) varied from 0.902 to 1 by receiver operating characteristics analysis]. miR-29c correlated with both estimated glomerular filtration rate ( r = 0.362; P &lt; 0.05) and degree of tubulointerstitial fibrosis ( r = −0.359; P &lt; 0.05) for CKD patients. Moreover, miRNA in exosome was decreased in mild fibrosis group compared with moderated to severe group. miR-29a and miR-29c could predict degree of tubulointerstitial fibrosis with AUC of 0.883 and 0.738 ( P &lt; 0.05). The sensitivity and specificity for distinguishing mild from moderate to severe fibrosis were 93.8 and 81.3% with the use of miR-29a and 68.8 and 81.3% for miR-29c. Overall, miR-29c in urinary exosome correlates with both renal function and degree of histological fibrosis, suggesting it as a novel, noninvasive marker for renal fibrosis.
[32]
Lv LL, Cao YH, Pan MM, et al. CD2AP mRNA in urinary exosome as biomarker of kidney disease[J]. Clin Chim Acta, 2014, 428: 26-31. DOI: 10.1016/j.cca.2013.10.003.
[33]
Xie Y, Jia Y, Cuihua X, et al. Urinary exosomal microRNA profiling in incipient type 2 diabetic kidney disease[J]. J Diabetes Res, 2017, 2017: 6978984. DOI: 10.1155/2017/6978984.
[34]
Barutta F, Tricarico M, Corbelli A, et al. Urinary exosomal microRNAs in incipient diabetic nephropathy[J]. PLoS One, 2013, 8(11): e73798. DOI: 10.1371/journal.pone.0073798.
[35]
Delić D, Eisele C, Schmid R, et al. Urinary exosomal miRNA signature in type ii diabetic nephropathy patients[J]. PLoS One, 2016, 11(3): e150154. DOI: 10.1371/journal.pone.0150154.
[36]
Li W, Yang S, Qiao R, et al. Potential Value of Urinary Exosome-Derived let-7c-5p in the Diagnosis and Progression of Type II Diabetic Nephropathy[J]. Clin Lab, 2018, 64(5): 709-718. DOI: 10.7754/Clin.Lab.2018.171031.
Diabetic nephropathy (DN) is a leading cause of death worldwide. Reliable biomarkers are demanded for the non-invasive diagnosis of DN. This study aims to investigate whether miRNA in urinary exosomes could serve as a potential biomarker in the diagnosis and progression of DN.Urine samples were collected from fifteen healthy controls, twenty type II diabetes without DN and twenty-eight type II patients with DN who underwent kidney biopsy. Differential centrifugation was used to isolate exosomes from urine samples and exosomes were confirmed by electron microscopy, nanoparticle tracking analysis (NTA), and western blot. MiRNAs including let-7c-5p, miR-29c-5p, miR-15b-5p, and RNU6 were detected by real-time quantitative polymerase chain reaction (RT-PCR).Electron microscopy and NTA verified a typical shape of exosomes with an average diameter of 100.7 nm, and western blot identified the presence of CD9, Alix, and TSG101 on exosomes. let-7c-5p was significantly upregulated in urinary exosomes of DN patients compared with controls (p < 0.05), while miR-29c-5p and miR-15b-5p were significantly downregulated compared with healthy controls (p < 0.05). let-7c-5p is correlated with both estimated glomerular filtration rate (r = -0.723, p < 0.001) and progression of DN. All three miRNAs, let-7c-5p, miR29c-5p, and miR-15b-5p, could predict DN with AUC of 0.818, 0.774, and 0.818, respectively.Urinary exosome-derived let-7c-5p is correlated with both renal function and progression of DN, suggesting it as a potential biomarker for DN.
[37]
Rossi L, Nicoletti MC, Carmosino M, et al. Urinary excretion of kidney aquaporins as possible diagnostic biomarker of diabetic nephropathy[J]. J Diabetes Res, 2017, 2017: 4360357. DOI: 10.1155/2017/4360357.
[38]
孙爱丽, 胡晓燕, 关广聚, 等. 尿胞外体亮氨酸氨基肽酶及二肽基肽酶在糖尿病肾病中的变化[J]. 中国病理生理杂志, 2011, 27(4): 775-778. DOI: 10.3969/j.issn.1000-4718.2011.04.029.
[39]
Zubiri I, Posada-Ayala M, Benito-Martin A, et al. Kidney tissue proteomics reveals regucalcin downregulation in response to diabetic nephropathy with reflection in urinary exosomes[J]. Transl Res, 2015, 166(5): 474-484. DOI: 10.1016/j.trsl.2015.05.007.
Diabetic nephropathy (DN) is a major complication of diabetes mellitus and the most frequent cause of end-stage renal disease. DN progresses silently and without clinical symptoms at early stages. Current noninvasive available markers as albuminuria account with severe limitations (late response, unpredictable prognosis, and limited sensitivity). Thus, it urges the discovery of novel markers to help in diagnosis and outcome prediction. Tissue proteomics allows zooming-in where pathophysiological changes are taking place. We performed a differential analysis of renal tissue proteome in a rat model of early DN by 2-dimensional differential gel electrophoresis and mass spectrometry. Confirmation was performed by Western blot, immunohistochemistry (IHC), and selected reaction monitoring (SRM). Rat urine samples were collected and exosomes were isolated from urine to evaluate if these microvesicles reflect changes directly occurring at tissue level. The protein showing maximum altered expression in rat tissue in response to DN was further analyzed in human kidney tissue and urinary exosomes. Regucalcin protein or senescence marker protein-30 (SMP30) (Swiss-Prot Q03336) was found to be strongly downregulated in DN kidney tissue compared with healthy controls. The same trend was observed in exosomes isolated from urine of control and DN rats. These data were further confirmed in a pilot study with human samples. IHC revealed a significant decrease of regucalcin in human kidney disease tissue vs control kidney tissue, and regucalcin was detected in exosomes isolated from healthy donors' urine but not from kidney disease patients. In conclusion, regucalcin protein expression is reduced in DN kidney tissue and this significant change is reflected in exosomes isolated from urine. Urinary exosomal regucalcin represents a novel tool, which should be explored for early diagnosis and progression monitoring of diabetic kidney disease. Copyright © 2015 Elsevier Inc. All rights reserved.
[40]
Katsuda T, Kosaka N, Takeshita F, et al. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles[J]. Proteomics, 2013, 13(10-11): 1637-1653. DOI: 10.1002/pmic.201200373.
Extracellular vesicles (EVs), membrane vesicles that are secreted by a variety of mammalian cell types, have been shown to play an important role in intercellular communication. The contents of EVs, including proteins, microRNAs, and mRNAs, vary according to the cell type that secreted them. Accordingly, researchers have demonstrated that EVs derived from various cell types play different roles in biological phenomena. Considering the ubiquitous presence of mesenchymal stem cells (MSCs) in the body, MSC-derived EVs may take part in a wide range of events. In particular, MSCs have recently attracted much attention due to the therapeutic effects of their secretory factors. MSC-derived EVs may therefore provide novel therapeutic approaches. In this review, we first summarize the wide range of functions of EVs released from different cell types, emphasizing that EVs echo the phenotype of their parent cell. Then, we describe the various therapeutic effects of MSCs and pay particular attention to the significance of their paracrine effect. We then survey recent reports on MSC-derived EVs and consider the therapeutic potential of MSC-derived EVs. Finally, we discuss remaining issues that must be addressed before realizing the practical application of MSC-derived EVs, and we provide some suggestions for enhancing their therapeutic efficiency.© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.
[41]
Jin J, Shi Y, Gong J, et al. Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte[J]. Stem Cell Res Ther, 2019, 10(1): 95. DOI: 10.1186/s13287-019-1177-1.
It is confirmed that adipose-derived stem cells (ADSCs) transplantation effectively relieves kidney fibrosis and type 2 diabetes disease in mice. Currently, exosome from urine-derived stem cells (USCs) can protect type 1 diabetes-mediated kidney injury and attenuate podocyte damage in diabetic nephropathy (DN). Exosome derived from USCs has evolved into the strategy for DN treatment, but the role of ADSCs-derived exosome (ADSCs-Exo) in DN remains unclear. The present study is aimed to investigate the therapeutic action and molecular mechanism of ADSCs-derived exosome on DN.ADSCs and exosome were authenticated by immunofluorescence and flow cytometry. Morphology and the number of exosome were evaluated by electron microscope and Nanosight Tracking Analysis (NTA), respectively. Cell apoptosis was assessed using flow cytometry. Podocyte autophagy and signaling transduction were measured by immunofluorescence and immunoblotting. Dual Luciferase Reporter assay was employed to detect the regulatory relationship between miR-486 and Smad1.ADSCs-Exo attenuated spontaneous diabetes by reducing levels of urine protein, serum creatinine (Scr), blood urea nitrogen (BUN), and podocyte apoptosis in mice. In in vitro experiment, ADSCs-Exo also reversed high glucose-induced decrease of cell viability and the increase of cell apoptosis in MPC5 cells. In terms of mechanism, ADSCs-Exo could enhance autophagy flux and reduce podocyte injury by inhibiting the activation of mTOR signaling in MPC5 and spontaneous diabetic mice. Eventually, we found that miR-486 was the key factors in ADSCs and in the process of ADSCs-Exo-mediated improvement of DN symptom in vivo and in vitro. miR-486 reduced Smad1 expression by target regulating Smad1 whose reduction could inhibit mTOR activation, leading to the increase of autophagy and the reduction of podocyte apoptosis.In conclusion, we illustrated that ADSCs-Exo vividly ameliorated DN symptom by enhancing the expression of miR-486 which led to the inhibition of Smad1/mTOR signaling pathway in podocyte. Possibly, ADSCs-Exo was used as a main therapeutic strategy for DN in future.
[42]
Jiang ZZ, Liu YM, Niu X, et al. Exosomes secreted by human urine-derived stem cells could prevent kidney complications from type I diabetes in rats[J]. Stem Cell Res Ther, 2016, 7: 24. DOI: 10.1186/s13287-016-0287-2.
Diabetic nephropathy is one of the most serious complications in patients with diabetes. At present, there are no satisfactory treatments available for diabetic nephropathy. Stem cells are currently the main candidates for the development of new treatments for diabetic nephropathy, as they may exert their therapeutic effects mainly through paracrine mechanisms. Exosomes derived from stem cells have been reported to play an important role in kidney injury. In this article, we try to investigate whether exosomes retrieved from urine stem cells could itself prevent diabetic nephropathy at an early stage in vivo and in vitro.
[43]
Li D, Wang N, Zhang L, et al. Mesenchymal stem cells protect podocytes from apoptosis induced by high glucose via secretion of epithelial growth factor[J]. Stem Cell Res Ther, 2013, 4(5): 103. DOI: 10.1186/scrt314.
The apoptosis and subsequent injury of podocytes plays a pathogenic role in diabetic nephropathy (DN). Mesenchymal stem cells (MSCs) are promising therapeutic cells for preventing apoptosis and reducing cellular injury. Our previous study found that MSCs could protect kidneys from diabetes-induced injury without obvious engraftment. So we evaluated the effects of human adipose-derived MSCs (hAd-MSCs) on podocytic apoptosis and injury induced by high glucose (HG) and the underlying mechanisms.We used flow cytometry, Western blot and confocal fluorescence microscopy to study podocytic apoptosis and injury induced by HG at 24 hours, 48 hours, and 72 hours in the presence or absence of MSC-conditioned medium (CM). An antibody-based cytokine array was used to identify the mediating factor, which was verified by adding the neutralizing antibody (NtAb) to block its function or adding the recombinant cytokine to the medium to induce its function.hAd-MSC-CM reduced podocytic apoptosis in a dose-dependent manner, decreased the expression of podocytic cleaved caspase-3, and prevented the reduced expression and maintained the normal arrangement of podocytic synaptopodin and nephrin. However, human embryonic lung cell (Wi38)-CM failed to ameliorate podocytic apoptosis or injury. Twelve cytokines with concentration ratios (MSC-CM/Wi38-CM) >10-fold were identified. Epithelial growth factor (EGF) was singled out for its known ability to prevent apoptosis. Recombinant human EGF (rhEGF) prevented podocytic apoptosis and injury similarly to hAd-MSC-CM but, upon blockade of EGF, the beneficial effect of hAd-MSC-CM decreased dramatically.hAd-MSCs prevent podocytic apoptosis and injury induced by HG, mainly through secreting soluble EG.
[44]
Nagaishi K, Mizue Y, Chikenji T, et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes[J]. Sci Rep, 2016, 6: 34842. DOI: 10.1038/srep34842.
Bone marrow-derived mesenchymal stem cells (MSCs) have contributed to the improvement of diabetic nephropathy (DN); however, the actual mediator of this effect and its role has not been characterized thoroughly. We investigated the effects of MSC therapy on DN, focusing on the paracrine effect of renal trophic factors, including exosomes secreted by MSCs. MSCs and MSC-conditioned medium (MSC-CM) as renal trophic factors were administered in parallel to high-fat diet (HFD)-induced type 2 diabetic mice and streptozotocin (STZ)-induced insulin-deficient diabetic mice. Both therapies showed approximately equivalent curative effects, as each inhibited the exacerbation of albuminuria. They also suppressed the excessive infiltration of BMDCs into the kidney by regulating the expression of the adhesion molecule ICAM-1. Proinflammatory cytokine expression (e.g., TNF-alpha) and fibrosis in tubular interstitium were inhibited. TGF-beta 1 expression was down-regulated and tight junction protein expression (e.g., ZO-1) was maintained, which sequentially suppressed the epithelial-to-mesenchymal transition of tubular epithelial cells (TECs). Exosomes purified from MSC-CM exerted an anti-apoptotic effect and protected tight junction structure in TECs. The increase of glomerular mesangium substrate was inhibited in HFD-diabetic mice. MSC therapy is a promising tool to prevent DN via the paracrine effect of renal trophic factors including exosomes due to its multifactorial action.
[45]
Liao W, Du Y, Zhang C, et al. Exosomes: the next generation of endogenous nanomaterials for advanced drug delivery and therapy[J]. Acta Biomater, 2019, 86: 1-14. DOI: 10.1016/j.actbio.2018.12.045.
Development of functional nanomaterials is of great importance and significance for advanced drug delivery and therapy. Nevertheless, exogenous nanomaterials have a great ability to induce undesired immune responses and nano-protein interactions, which may result in toxicity and failure of therapy. Exosomes, a kind of endogenous extracellular vesicle (40-100 nm in diameter), are considered as a new generation of a natural nanoscale delivery system. Exosomes secreted by different types of cells carry different signal molecules (such as RNAs and proteins) and thus have a great potential for targeted drug delivery and therapy. Herein, we provide comprehensive understanding of the properties and applications of exosomes, including their biogenesis, biofunctions, isolation, purification, and drug loading, and typical examples in drug delivery and therapy. Furthermore, their advantages compared to other nanoparticles and potential in tumor immunotherapy are also discussed. STATEMENT OF SIGNIFICANCE: Exosomes, a kind of endogenous extracellular vesicle, have emerged as a novel and attractive endogenous nanomaterial for advanced drug delivery and targeted therapy. Exosomes are secreted by many types of cells and carry some unique signals obtained from their parental cells. Furthermore, the liposome-like structure allows exosomes to load various drugs. Hence, the potential of exosomes in drug delivery, tumor targeted therapy, and immunotherapy has been investigated in recent years. On the basis of their endogenous features and multifunctional properties, exosomes are of great significance and interest for the development of future medicine and pharmaceuticals.Copyright © 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
[46]
Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery[J]. J Control Release, 2015, 219: 396-405. DOI: 10.1016/j.jconrel.2015.07.030.
[47]
Tang TT, Lv LL, Wang B, et al. Employing macrophage-derived microvesicle for kidney-targeted delivery of dexamethasone: an efficient therapeutic strategy against renal inflammation and fibrosis[J]. Theranostics, 2019, 9(16): 4740-4755. DOI: 10.7150/thno.33520.
PDF(968 KB)

2049

Accesses

0

Citation

Detail

Sections
Recommended

/