Research Progress on the Role of Mesenchymal Stem Cell Extracellular Vesicles in Lung Diseases

WANG Ze-hua, ZHANG Li-yun, MA Chun-yan

China Biotechnology ›› 2023, Vol. 43 ›› Issue (5) : 76-84.

PDF(663 KB)
PDF(663 KB)
China Biotechnology ›› 2023, Vol. 43 ›› Issue (5) : 76-84. DOI: 10.13523/j.cb.2212035

Research Progress on the Role of Mesenchymal Stem Cell Extracellular Vesicles in Lung Diseases

Author information +
History +

Abstract

Extracellular vesicles (EVs) are lipid vesicles naturally secreted by cells. They play an important role in communication of information in physiological and pathological processes. Mesenchymal stem cells are pluripotent stromal stem cells from a wide range of sources. The potential of mesenchymal stem cell regeneration and its ability of immunomodulation have shown great promise in the repair in and treatment of lung diseases. Mesenchymal stromal cells-EVs (MSCs-EVs) have the same functional characteristics as MSCs, and many active factors carried by MSCs-EVs have shown good therapeutic effects in lung tissue, lung microenvironment and lung diseases. In this paper, the biological characteristics of MSCs and MSCs-EVs were summarized, and the mechanism and clinical application of MSCs-EVs in pulmonary diseases were discussed.

Key words

Mesenchymal stem cell / Extracellular vesicles / Lung disease / Treatment

Cite this article

Download Citations
Ze-hua WANG, Li-yun ZHANG, Chun-yan MA. Research Progress on the Role of Mesenchymal Stem Cell Extracellular Vesicles in Lung Diseases[J]. China Biotechnology, 2023, 43(5): 76-84 https://doi.org/10.13523/j.cb.2212035
吸烟、创伤、空气污染及病原体感染等多种因素都会引起气道和肺部结构出现功能障碍,引发多种类型肺部疾病的发生。近年来,肺部疾病的发病率和死亡率急剧增高,已成为全球主要公共卫生问题之一。在最近十几年里,干细胞疗法在治疗肺部疾病和组织损伤方面得到了广泛关注。在各种类型的干细胞中,间充质干细胞(mesenchymal stromal cell,MSCs)因其潜在的再生和免疫调节作用在肺部疾病治疗方面引起关注[1]。尽管MSCs的作用机制尚未完全阐明,但在肺部疾病中局部或全身注射MSCs具有促进生物活性物质释放、激活相关内源性修复途径和重编程途径,影响肺部环境的炎症,并对肺部重塑过程进行调节,达到修复和治疗肺组织的目的[2]
细胞外囊泡(extracellular vesicles,EVs)是细胞在多种生理状态释放的膜结合球形结构,携带蛋白质、核酸、脂质等生物活性分子,在细胞通信中发挥重要作用。体内或体外培养的MCSs在生长过程中会分泌大量细胞因子,其中EV所占比率最高。MSCs-EV可通过递送遗传信息、蛋白质、活性因子等物质介导MSCs与靶细胞之间的相互作用,在心血管疾病、慢性肾脏疾病、呼吸系统疾病和癌症治疗中引起广泛关注[3]。MSCs-EV具有类似MSCs的治疗效果,也能避免细胞疗法在疾病中的局限性,如细胞群体的异质性、肿瘤发生、栓塞、MSCs的细胞质量等[4]。近年来,越来越多的研究集中在MSC-EVs在慢性呼吸系统疾病中的治疗作用上。本文综述了MSCs和MSCs-EV的特性,以及MSCs-EV在多种肺部疾病中的作用,为预防和治疗呼吸系统疾病提供新视角。

1 间充质干细胞

MSCs是一类具有自我更新、再生、分化和修复功能,来源于中胚层的多能干细胞[5-6]。MSCs在1971年由Friedenstein等[7]从豚鼠骨髓中发现,Caplan[8]在1991年将其命名为间充质干细胞,1998年Majumdar等[9]进一步证明了这些细胞存在于包括人类在内的其他物种。MSCs在人体组织中分布广泛,通常从骨髓、脂肪组织、牙髓、胎盘、脐带、肝脏、滑膜和其他组织分离获得[6]。不同组织的MSCs具有相似形态和表型特征,但决定MSCs特定功能的是其细胞起源[10]。在机体免疫系统中MSCs可以调节各类免疫细胞的激活和功能,但这种免疫调节能力不是自身具有的,而是取决于MSCs存在的炎症环境。近年来,由于MSCs相对容易从各种组织中提取并在体外培养,且具有免疫调节、促进细胞存活、高分化潜力、归巢能力等优点,在肺部疾病、神经系统疾病、造血性疾病、心血管疾病等多种疾病的研究潜力得到广泛关注[11]。此外,MSCs在生长存活过程中可以产生多种活性物质如生长因子、细胞因子、趋化因子,也间接参与组织损伤再生的细胞外囊泡。现在,越来越多的证据表明MSCs的多种生理功能与本身的旁分泌成分有关,而细胞外囊泡作为MSCs分泌最多也最重要的成分随之成为研究热点[12]

2 细胞外囊泡及间充质干细胞胞外囊泡

EVs是细胞自然释放颗粒的总称,在机体内部细胞通信机制及各种生理过程中充当媒介,发挥重要作用[13]。EVs为球形,其本质为脂质双层结构,几乎所有类型细胞和生物都普遍分泌,目前已从细培养上清液和多种组织液中提取,包括血浆、唾液、尿液、胆汁、羊水、母乳、精液和腹水等[14]。根据颗粒大小、生物发生和分泌机制的不同,细胞外囊泡通常分为外泌体、微泡和凋亡小体[15](图1)。外泌体和微泡是最早研究的EV类型,外泌体直径为30~150 nm,由多胞体(MVB)与质膜融合向外释放[16]。与外泌体相比,微泡直径为100~1 000 nm,由质膜向外出芽分裂形成;而凋亡小体直径为500~4 000 nm,是细胞凋亡过程中细胞起泡产生[17-18]。不同来源EV具有很大的异质性,其包裹的核酸、蛋白质、脂质和代谢物等可以反映不同病理条件下机体的代谢状态和亲代细胞的生理功能,临床诊断具有极大潜力[19]。迄今为止,从MSC-EVs已经鉴定出5 000多种蛋白质,这些蛋白质的调控功能包括控制自我更新、分化、迁移、增殖、调控免疫反应等。MSCs-EV也含有许多黏附分子,包括CD44、CD29、α4-整合素和α5-整合素等,这些分子有助于对MSCs-EVs的鉴定[20]。目前,已发现MSCs-EV的miRNA有150多种,这些miRNA的功能通常与细胞凋亡、肿瘤发生、免疫反应、血管生成和生物体发育有关[21]。近年来,在肺部疾病的防治方面MSCs-EV调控作用展现出优良前景。
Fig.1 Classification of extracellular vesicles

图1 细胞外囊泡分类

Full size|PPT slide

3 MSCs-EV作为药物运输的理想载体

肺是呼吸系统的主要器官,由局部基质细胞和常驻免疫细胞之间的通信维持肺内稳定。EV作为细胞间通信的重要载体在维持肺内微环境稳态方面发挥重要作用[22]。虽然从生物体液提取的EV是不同疾病诊断的生物标志物,但细胞来源的EV仍是药物递送的首选。几乎所有细胞都会分泌EV,最为常见的细胞包括免疫细胞、癌细胞和MSCs。与其他细胞来源的EV相比,MSCs是分泌EV最多的细胞,MSCs-EV具有和MSCs同样的免疫抑制活性和免疫调节特性[23]。多项临床试验表明,MSCs-EV具有良好的治疗效果,并且在不同疾病动物模型中具有耐受性,且无明显不良反应[24]。此外,MSCs-EV还继承亲本细胞的内在肿瘤性,其复杂表面蛋白为靶向外源配体和通过其他表面修饰手段增强靶向能力提供了工程化方向。MSCs-EV在生物工程化改造方面也表现出很高的灵活性,在储存过程中展现出良好的稳定性。药物加载和表面修饰是MSCs-EV工程化最常用的手段,这些方式不但避免MSCs-EV被吞噬细胞吞噬增强靶向性,也极大提高MSCs-EV运输药物的效率。与传统纳米载体相比,MSCs-EV是天然的内源性载体,具有更高的生物相容性和更低的免疫原性[25]。在给药方面,通过MSCs-EV给药可以在初次治疗时降低患者对药物的抵抗性,大大提高药物的有效性[26]。在穿过血脑屏障和血-视网膜等生物屏障中,MSCs-EV可以自由通过,具有更好的通透性,在治疗眼和中枢神经系统疾病方面显示出光明的前景。

4 间充质干细胞来源细胞外囊泡与肺部疾病

4.1 支气管肺发育不良

支气管肺发育不良(bronchopulmonary dysplasia,BPD)是经常发生于新生儿或早产儿的一种慢性呼吸系统疾病,通常采取药物、呼吸管理、营养支持等方式对BPD进行治疗,但这些方法的有效性及利弊不明确,目前尚未存在改善BPD患者的有效方法[27]。在MSCs研究中,通常将新生小鼠暴露于高氧(75%)微环境来模拟BPD病理模型。Willis等[28]首次对MSCs-EV进行研究,实验表明将MSCs来源外泌体以8.5×108颗粒/50 μL剂量静脉给药可增强巨噬细胞表型和外周肺动脉重塑,改善高氧条件的新生小鼠肺室间隔纤维化和肺部发育,恢复肺结构。肿瘤坏死因子刺激蛋白-6(tumor necrosis factorαstimulating gene-6 protein,TSG-6)是MSCs发挥免疫抑制作用的关键因子,一般受肿瘤坏死因子α(TNF-α)和白介素-1β(IL-1β)等炎症因子的影响而产生,不但可以诱导巨噬细胞从促炎M1型向抗炎M2型转变,还可以通过MSC分泌的外泌体传递TSG-6因子恢复肺泡毛细血管渗透压,并减少肺泡空心化,缓解高氧诱导的BPD小鼠[29]。绒毛膜羊膜炎是BPD发生的主要因素,由内毒素诱导的新生小鼠BPD模型中,骨髓来源MSCs-EV治疗可减少促炎因子如趋化因子I(IP-10)的表达,增强抗炎因子如白介素-4(IL-4)、白介素-13(IL-13)的表达,并逆转BPD小鼠模型中的胎盘血管发育和产后远端肺生长[30]。血管内皮生长因子(vascular endothelial growth factor,VEGF)也是BPD治疗中研究最多的方向,MSCs-EV所携带的VEGF是一种关键的旁分泌因子,在减轻新生大鼠的高氧BPD中起着重要作用[31]。此外,多种方式MSCs-EV给药在调控巨噬细胞极化、改善肺泡形成、血管生成及降低胶原密度方面具有重要作用,也在BPD治疗方面具有极大潜力。

4.2 急性呼吸窘迫综合征

急性呼吸窘迫综合征(acute respiratory distress syndrome,ARDS)是一种发病率和死亡率都较高,由脓毒症、肺炎和外伤引起的急性炎症性肺部疾病[32]。MSCs分泌的多种因子在恢复上皮和内皮细胞通透性、促进巨噬细胞吞噬、下调急性炎症、改善肺泡液清除率等方面发挥重要功能[33]。在多种因素诱导的急性肺损伤模型中,MSCs-EV已被证明对ARDS产生有益作用。在甲型流感H5N1诱导的肺损伤中,与脐带来源的MSCs作用相比,MSCs-EV在降低肺泡液清除率和肺泡上皮细胞的蛋白质通透性方面作用更明显。另外,MSCs-EV也可进入上皮细胞,在其他流感诱导的肺损伤模型中能够抑制病毒复制、肺泡细胞凋亡和血凝活性[34]。肺内皮细胞和上皮细胞是ARDS中受损伤的主要细胞类型,已经广泛进行了MSCs-EV对这些细胞修复潜力的研究。Hu等[35]发现MSCs-EV能够将血管生成素-1的mRNA转移到受损伤的内皮细胞来恢复人肺血管内皮细胞,防止肌动蛋白“应激纤维”的形成。目前,仅在少部分研究中进行MSCs-EV对肺泡上皮细胞生物学功能的探讨。MSCs-EV携带的miRNA在减轻ARDS中同样发挥重要作用。据报道,MSCs-EV通过传递miR-21-5p、miR-30b-3p、miR-100、miR-145a、miR-146a来减轻炎症反应,也通过转运miR-27a-3p、miR-146a来调节巨噬细胞极化[36-41]。线粒体的能量调节在ARDS的发病机制中也起着关键作用,MSCs-EV可以向巨噬细胞转运功能性线粒体,通过增强氧化磷酸化来调节巨噬细胞极化,改善上皮细胞的线粒体功能,从而在ARDS模型中促进伤口闭合[42]。目前预防ARDS后肺纤维化是改善长期预后的最佳方法,因此需要研究MSCs-EV在预防或逆转ARDS后肺纤维化等方面的潜在功能。

4.3 特发性肺纤维化

特发性肺纤维化是一种慢性、间质性肺部疾病,成纤维细胞异常增殖和细胞外基质过度沉积是其主要病理特征[43]。其病理因素与遗传因素和环境因素密切相关,但仍无明确病因和有效疗法[44]。在预防和治疗博莱霉素诱导的肺纤维化中,静脉注射MSCs-EV可改善肺形态结构并减少胶原沉积,调节巨噬细胞经典和非经典表型转变,使肺泡巨噬细胞的数量转向正常[45]。在二氧化硅诱导小鼠硅肺纤维化中,MSCs-EV可以调节肺功能,抑制硅肺纤维化的进程[46]。此外,骨髓MSCs-EV也同样抑制纤维组织中Wnt/β-catenin相关蛋白质的表达,逆转上皮间质转化缓解二氧化硅诱导的肺纤维化[47]。MSCs-EV运输中的miRNA抗纤维化作用也成为研究热点。Sun等[48]研究表明,来自月经血的MSCs-EV通过传递miR-let-7到肺泡上皮细胞中调节活性氧、线粒体DNA损伤和NLRP3炎性小体激活来缓解肺纤维化。同样脂肪来源MSCs-EV向上皮细胞转移miR-let-7,通过靶向TGF-βRI基因,降低ROS水平和炎症水平,抑制PM2.5诱导的肺纤维化[49]。成纤维细胞增殖在肺纤维化中起着重要作用,Wan等[50]研究表明骨髓来源MSCs-EV通过miR-29-3p下调成纤维细胞中FZD6的表达抑制成纤维细胞增殖进而抑制肺纤维化发生。另有一项研究同样发现,骨髓来源MSCs-EV中的miR-186通过抑制SRY相关的HMG盒转录因子4(SOX4)和下游基因Dickkopf-1(DKK1)的表达来阻断成纤维细胞活化缓解肺纤维化[51]。MSCs-EV还可以转移蛋白质缓解肺纤维化,人基质来源MSCs-EV能够向人基底支气管细胞转移锌指激活剂来激活囊性纤维化跨膜传导调节因子(CFTR)来治疗肺纤维化[52]

4.4 慢性阻塞性肺病

慢性阻塞性肺病(chronic obstructive pulmonary disease,COPD)是一种气体交换功能障碍,气道和肺泡受损产生炎症反应的进行性肺部疾病[53]。香烟烟雾是引发COPD的首要因素,香烟烟雾可诱导中性粒细胞、淋巴细胞和巨噬细胞数量增加,释放促炎因子表达引起肺部炎症[54]。在香烟烟雾诱导的COPD小鼠模型中,MSCs来源外泌体可以明显降低巨噬细胞和嗜中性粒细胞的数量,减少肿瘤坏死因子-α(tumor necrosis factor-α,TNF-α)和IL-1β的产生,恢复O2饱和度,改善肺功能和减弱的气道炎症[55]。成纤维细胞生长因子-2(fibroblast growth factor 2,FGF-2)是一种具有再生作用的因子,对肺部发育具有重要作用。与MSCs来源的天然外泌体相比,MSCs来源人工合成纳米囊泡同样存在相似的表面标志物、生长因子和FGF-2。此外,人工纳米囊泡显示出更强大的再生效果,其主要通过激活FGF-2信号通路来产生,表明人工纳米囊泡具有极大经济优势,临床适用于COPD患者[56]。线粒体功能失调也是促进COPD发生的重要起因,MSCs-EV还可以通过调控靶细胞失调的线粒体而在COPD中发挥治疗功效。Maremanda等[57]证明在香烟烟雾诱导的COPD模型中,小鼠MSCs来源外泌体可以修饰支气管上皮细胞中的线粒体基因,缓解线粒体功能障碍和炎症反应。MSCs-EV治疗作为一个前沿领域,通过调节慢性炎症,抑制肺气肿和恢复功能失调的线粒体,在治疗COPD中具有令人期望的前景。

4.5 硅肺

硅肺是一种职业性慢性呼吸系统疾病,由长时间吸入结晶二氧化硅粉尘引起肺部炎症,导致进行性和不可逆的肺纤维化,缺乏特异性药物治疗并可能增加肺结核的发病率[58]。在二氧化硅诱导的硅肺模型中,通过移植MSCs可降低纤维化标志蛋白和胱天蛋白酶-3的表达,增加抗凋亡蛋白Bcl-2和促凋亡蛋白Bax的比值和上皮标志蛋白的含量,降低胶原蛋白III和纤连蛋白的mRNA水平以及转化生长因子-β(transforming growth factor-β,TGF-β)和羟脯氨酸的含量,对二氧化硅诱导的肺纤维化产生抑制作用[59]。Choi等[60]首次证明人骨髓MSCs-EV能减轻小鼠硅肺肺纤维化模型中炎症细胞的流入和胶原沉积,虽然MSCs-EV对硅肺具有有益作用,但其治疗效果远不如MSC移植。骨髓MSCs-EV通过携带逮捕素结构域的蛋白质1将去极化的线粒体靶向质膜调节细胞内氧化应激反应,随后这些EV被吞噬、吸收而增强生物能量,并在巨噬细胞融合过程中被重新利用[61]。此外,MSCs-EV所包含的miRNA可抑制Toll样受体信号转导来抑制巨噬细胞活化,从而使巨噬细胞对摄入的线粒体脱敏。在硅肺小鼠模型中,通过气管滴注脂肪MSCs-EV可减少胶原沉积、肉芽肿大小和巨噬细胞流入[62]。目前,尽管MSCs-EV在硅肺中展现优良的治疗前景,但具体机制仍有待后续研究。

4.6 哮喘

哮喘是一种发生在成人和儿童体内的慢性炎症性的肺部疾病[63]。在开发和研究新的治疗方法过程中,MSCs在缓解和治疗哮喘的主要病理特征方面具有极大潜力。单次注射MSCs可减少嗜酸性粒细胞、淋巴细胞、总蛋白质、H2O2及肺泡灌洗液中(BALF)白介素因子(IL-5、IL-13和IL-17a)的表达水平,减轻肺部炎症浸润、胶原蛋白沉积及黏液产生[64]。近年来,有关MSCs-EV治疗过敏性哮喘的研究已有进展。MSCs-EV能促进外周血单核细胞IL-10和TGF-β1表达,进而调节T细胞增殖和免疫能力,这些研究结果表明MSCs-EV对哮喘的治疗潜力[65]。脂肪来源的MSCs和MSCs-EV能有效降低肺组织和BALF中的嗜酸性粒细胞数量,减少炎症并调节气道重塑[66]。在抑制炎性细胞浸润、黏液分泌、气道高反应性、下调T细胞和调控先天淋巴细胞功能方面,MSCs-EV所包括的miR-146a-5p已被证明介导上述功能,使先天淋巴细胞能显著预防过敏性气道炎症[67]。缺氧环境可促使MSCs释放更多EV,人脐带MSCs-EV显著改善哮喘小鼠的BALF总细胞、嗜酸性粒细胞和促炎介质,并且MSCs在缺氧条件下产生的EVs比正常条件下的EVs治疗哮喘的效果更显著[68]

4.7 肺动脉高压

肺动脉高压(pulmonary arterial hypertension,PAH)是一种罕见的进行性疾病,是指肺循环的血流动力学改变,其中肺动脉压为>25 mmHg[69]。尽管在阐明PAH的治疗及其分子机制方面取得重大进展,但PAH对大多数常规药物治疗仍然无效。MSCs-EV在预防和治疗PAH中展现出优良前景。为研究MSCs-EV对肺血管系统的影响,Lee等[70]对缺氧条件诱导的PAH小鼠模型采取静脉注射MSCs-EV的方式,可抑制肺巨噬细胞流入,促进多种炎症因子表达,并抑制血管重塑,防止右心室肥大而导致PAH。缺氧是参与肺血管系统对缺氧反应的关键因素,MSCs-EV通过抑制缺氧诱导的转录因子3的激活和上调miRNA簇的miR-17,增加miR-204的肺内表达水平,而miR-204是一种已知在人类肺动脉高压中受到抑制的miRNA。在苦百合碱诱导的PAH大鼠模型中,静脉注射MSCs和MSCs-EV可以改善平均肺动脉压和平均右心室压,也可以降低PAH大鼠右心室肥大及肺小动脉面积指数和厚度指数[71]。此外,骨髓MSCs-EV不仅可以预防苦百合碱诱导的PAH发生,还可以逆转肺高压变化,包括右心室肥大和肺血管重塑,这些变化取决于MSCs-EV所携带的miRNA种类[72]。在这项研究中,还发现相比MSCs来源微泡,MSCs所分泌外泌体具有更好的治疗效果,与从健康小鼠血浆中收获的外泌体相比,这种差异部分归因于MSCs外泌体富含抗炎和抗增殖miRNA,包括miR-34a、miR-122、miR-124和miR-127。另外,脂肪来源MSCs外泌体包裹的miR-191通过抑制骨形态发生蛋白受体2的表达来恢复久效吡咯诱导的PAH[73]。在缺氧诱导的PAH小鼠模型中,MSCs来源外泌体还可以通过上调丙酮酸脱氢酶和谷氨酸脱氢酶1的表达,改善线粒体功能障碍缓解PAH[74]

5 总结与展望

在全球范围内,肺部呼吸系统疾病仍然严重威胁数百万人的健康,因此迫切需要开发新的预防或治疗方法去面对呼吸系统疾病的挑战。MSCs-EV作为一种全新的无细胞疗法,在治疗肺部疾病等方面具有比MSCs更安全、更有效、更方便的优势,有希望替代MSCs移植成为全新治疗方案。首先,MSCs-EV有高度稳定性,适合长期储存,且无须添加有潜在毒性的冷冻保存剂。其次,MSC-EVs可直接将功能蛋白和miRNA转移到受体细胞,在细胞间通信中发挥更强大的信号转导功能。最后,MSCs-EV无任何异源风险,同种异体应用后没有免疫反应,也可以避免MSCs治疗的潜在致瘤性风险。
虽然MSCs-EV在肺部疾病中的治疗潜力得到广泛关注,但对MSCs-EV的研究仍处于起步阶段,在临床治疗中仍存在诸多问题。现阶段,大部分体内和体外实验所采用的MSCs和MSCs-EV都未经处理。MSCs的大规模培养在临床研究依然是一大难题,需保证MSCs培养传代过程中无污染和生理状态一致。EV的提取、分离和纯化仍未有明确的标准规范,导致获得的EV存在极大异质性特征,需要统一EV的分离标准,并且开发新的提取分离方法确保纯度和质量。MSCs-EV发挥功效的精准货物分子仍有待确定,并且可能因细胞类型、培养条件以及提取和纯化方法等因素而异,对MSCs-EV的功能作用有潜在影响。由于MSCs是从各种供体中收获的,因此需要探索了解供体的特征如何改变EV成分及功能。MSCs-EV的功能化策略仍有局限性,缺乏系统性比较和评价体系。目前有关药物加载方法较多,药物加载效率决定MSCs-EV治疗的成败,因此要对不同药物建立特定的有针对性的加载策略,确保药物尽最大程度发挥功效。在多种肺部疾病中MSCs-EV的给药方式、给药剂量、给药次数均有所不同,还需后续大规模临床研究统一确定。目前,MSCs-EV的治疗潜力在肺部疾病得到广泛研究,但具体是MSCs-EV的哪种成分发挥功效以及具体机制是什么尚未明确,因此需要进一步深入研究讨论。综上所述,虽然MSCs-EV仍存在诸多尚未解决的问题,但随着生命科学与生物技术的不断进步,这些问题有望逐一被解决,MSCs-EV作为一种非细胞的治疗方式有着更为广阔的前景。

References

[1]
Cruz F F, Rocco P R M. The potential of mesenchymal stem cell therapy for chronic lung disease. Expert Review of Respiratory Medicine, 2020, 14(1): 31-39.
: Mesenchymal stem/stromal cells (MSCs) have been shown to improve lung function and survival in chronic inflammatory lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension (PAH), and silicosis.: This review covers rationale for the use of MSC therapy, along with preclinical studies and clinical trials with MSC therapy in chronic lung diseases.: MSC therapy holds promise for the treatment of chronic lung diseases, mainly when administered at early stages. In clinical trials, MSC administration was safe, but associated with limited effects on clinical outcomes. Further studies are required to elucidate unresolved issues, including optimal MSC source and dose, route of administration, and frequency (single. multiple-dose regimens). A better understanding of the mechanisms of MSC action, local microenvironment of each disease, and development of strategies to potentiate the beneficial effects of MSCs may improve outcomes.
[2]
Guo H Y, Su Y, Deng F. Effects of mesenchymal stromal cell-derived extracellular vesicles in lung diseases: current status and future perspectives. Stem Cell Reviews and Reports, 2021, 17(2): 440-458.
Mesenchymal stromal cells (MSCs) as a kind of pluripotent adult stem cell have shown great therapeutic potential in relation to many diseases in anti-inflammation and regeneration. The results of preclinical experiments and clinical trials have demonstrated that MSC-derived secretome possesses immunoregulatory and reparative abilities and that this secretome is capable of modulating innate and adaptive immunity and reprograming the metabolism of recipient cells via paracrine mechanisms. It has been recognized that MSC-derived secretome, including soluble proteins (cytokines, chemokines, growth factors, proteases), extracellular vesicles (EVs) and organelles, plays a key role in tissue repair and regeneration in bronchopulmonary dysplasia, acute respiratory distress syndrome (ARDS), bronchial asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension, and silicosis. This review summarizes the known functions of MSC-EV modulation in lung diseases, coupled with the future challenges of MSC-EVs as a new pharmaceutical agent. The identification of underlying mechanisms for MSC-EV might provide a new direction for MSC-centered treatment in lung diseases.Graphical abstract.
[3]
de Abreu R C, Fernandes H, da Costa Martins P A, et al. Native and bioengineered extracellular vesicles for cardiovascular therapeutics. Nature Reviews Cardiology, 2020, 17(11): 685-697.
[4]
Shi M M, Yang Q Y, Monsel A, et al. Preclinical efficacy and clinical safety of clinical-grade nebulized allogenic adipose mesenchymal stromal cells-derived extracellular vesicles. Journal of Extracellular Vesicles, 2021, 10(10): e12134.
[5]
Yang C J, Sun J M, Tian Y P, et al. Immunomodulatory effect of MSCs and MSCs-derived extracellular vesicles in systemic lupus erythematosus. Frontiers in Immunology, 2021, 12: 714832.
Systemic lupus erythematosus (SLE) is a common autoimmune connective tissue disease with unclear etiology and pathogenesis. Mesenchymal stem cell (MSC) and MSC derived extracellular vesicles (EVs) play important roles in regulating innate and adaptive immunity, which are involved in many physiological and pathological processes and contribute to the immune homeostasis in SLE. The effects of MSCs and EVs on SLE have been drawing more and more attention during the past few years. This article reviews the immunomodulatory effects and underlying mechanisms of MSC/MSC-EVs in SLE, which provides novel insight into understanding SLE pathogenesis and guiding the biological therapy.
[6]
Aravindhan S, Ejam S S, Lafta M H, et al. Mesenchymal stem cells and cancer therapy: insights into targeting the tumour vasculature. Cancer Cell International, 2021, 21(1): 158.
A crosstalk established between tumor microenvironment and tumor cells leads to contribution or inhibition of tumor progression. Mesenchymal stem cells (MSCs) are critical cells that fundamentally participate in modulation of the tumor microenvironment, and have been reported to be able to regulate and determine the final destination of tumor cell. Conflicting functions have been attributed to the activity of MSCs in the tumor microenvironment; they can confer a tumorigenic or anti-tumor potential to the tumor cells. Nonetheless, MSCs have been associated with a potential to modulate the tumor microenvironment in favouring the suppression of cancer cells, and promising results have been reported from the preclinical as well as clinical studies. Among the favourable behaviours of MSCs, are releasing mediators (like exosomes) and their natural migrative potential to tumor sites, allowing efficient drug delivering and, thereby, efficient targeting of migrating tumor cells. Additionally, angiogenesis of tumor tissue has been characterized as a key feature of tumors for growth and metastasis. Upon introduction of first anti-angiogenic therapy by a monoclonal antibody, attentions have been drawn toward manipulation of angiogenesis as an attractive strategy for cancer therapy. After that, a wide effort has been put on improving the approaches for cancer therapy through interfering with tumor angiogenesis. In this article, we attempted to have an overview on recent findings with respect to promising potential of MSCs in cancer therapy and had emphasis on the implementing MSCs to improve them against the suppression of angiogenesis in tumor tissue, hence, impeding the tumor progression.
[7]
Friedenstein A J, Chailakhjan R K, Lalykina K S. The development of fibroblast colonies in monolayer cultures of Guinea-pig bone marrow and spleen cells. Cell and Tissue Kinetics, 1970, 3(4): 393-403.
[8]
Caplan A I. Mesenchymal stem cells. Journal of Orthopaedic Research: Official Publication of the Orthopaedic Research Society, 1991, 9(5): 641-650.
[9]
Majumdar M K, Thiede M A, Mosca J D, et al. Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. Journal of Cellular Physiology, 1998, 176(1): 57-66.
Mesenchymal stem cells (MSCs) are a population of pluripotent cells within the bone marrow microenvironment defined by their ability to differentiate into cells of the osteogenic, chondrogenic, tendonogenic, adipogenic, and myogenic lineages. We have developed methodologies to isolate and culture-expand MSCs from human bone marrow, and in this study, we examined the MSC's role as a stromal cell precursor capable of supporting hematopoietic differentiation in vitro. We examined the morphology, phenotype, and in vitro function of cultures of MSCs and traditional marrow-derived stromal cells (MDSCs) from the same marrow sample. MSCs are morphologically distinct from MDSC cultures, and flow cytometric analyses show that MSCs are a homogeneous cell population devoid of hematopoietic cells. RT-PCR analysis of cytokine and growth factor mRNA in MSCs and MDSCs revealed a very similar pattern of mRNAs including IL-6, -7, -8, -11, -12, -14, and -15, M-CSF, Flt-3 ligand, and SCF. Steady-state levels of IL-11 and IL-12 mRNA were found to be greater in MSCs. Addition of IL-1alpha induced steady-state levels of G-CSF and GM-CSF mRNA in both cell preparations. In contrast, IL-1alpha induced IL-1alpha and LIF mRNA levels only in MSCs, further emphasizing phenotypic differences between MSCs and MDSCs. In long-term bone marrow culture (LTBMC), MSCs maintained the hematopoietic differentiation of CD34+ hematopoietic progenitor cells. Together, these data suggest that MSCs represent an important cellular component of the bone marrow microenvironment.
[10]
Wu R Q, Fan X L, Wang Y, et al. Mesenchymal stem cell-derived extracellular vesicles in liver immunity and therapy. Frontiers in Immunology, 2022, 13: 833878.
Mesenchymal stem cells (MSCs), as the most common cell source for stem cell therapy, play an important role in the modulation of innate and adaptive immune responses and have been widely used in clinical trials to treat autoimmune and inflammatory diseases. Recent experimental and clinical studies have shown that MSC-derived extracellular vesicles (MSC-EVs) can inhibit the activation and proliferation of a variety of proinflammatory cells, such as Th1, Th17 and M1 macrophages, reducing the secretion of proinflammatory cytokines, while promoting the proliferation of anti-inflammatory cells, such as M2 macrophages and Tregs, and increasing the secretion of anti-inflammatory cytokines, thus playing a role in immune regulation and exhibiting immunomodulatory functions. Besides MSC-EVs are more convenient and less immunogenic than MSCs. There is growing interest in the role of MSC-EVs in liver diseases owing to the intrinsic liver tropism of MSC-EVs. In this review, we focus on the immunomodulatory effects of MSC-EVs and summarize the pivotal roles of MSC-EVs as a cell-free therapy in liver diseases, including NAFLD, AIH, acute liver failure, liver fibrosis and hepatic ischemia–reperfusion injury. Moreover, we provide a concise overview of the potential use and limits of MSC-EVs in clinical application.
[11]
Keshtkar S, Azarpira N, Ghahremani M H. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Research & Therapy, 2018, 9(1): 63.
[12]
Rezaie J, Nejati V, Mahmoodi M, et al. Mesenchymal stem cells derived extracellular vesicles: a promising nanomedicine for drug delivery system. Biochemical Pharmacology, 2022, 203: 115167.
[13]
van Niel G, Carter D R F, Clayton A, et al. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nature Reviews Molecular Cell Biology, 2022, 23(5): 369-382.
Extracellular vesicles (EVs) are increasingly recognized as important mediators of intercellular communication. They have important roles in numerous physiological and pathological processes, and show considerable promise as novel biomarkers of disease, as therapeutic agents and as drug delivery vehicles. Intriguingly, however, understanding of the cellular and molecular mechanisms that govern the many observed functions of EVs remains far from comprehensive, at least partly due to technical challenges in working with these small messengers. Here, we highlight areas of consensus as well as contentious issues in our understanding of the intracellular and intercellular journey of EVs: from biogenesis, release and dynamics in the extracellular space, to interaction with and uptake by recipient cells. We define knowledge gaps, identify key questions and challenges, and make recommendations on how to address these.© 2022. Springer Nature Limited.
[14]
Yáñez-Mó M, Siljander P R M, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles, 2015, 4(1): 27066.
[15]
van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nature Reviews Molecular Cell Biology, 2018, 19(4): 213-228.
Extracellular vesicles are a heterogeneous group of cell-derived membranous structures comprising exosomes and microvesicles, which originate from the endosomal system or which are shed from the plasma membrane, respectively. They are present in biological fluids and are involved in multiple physiological and pathological processes. Extracellular vesicles are now considered as an additional mechanism for intercellular communication, allowing cells to exchange proteins, lipids and genetic material. Knowledge of the cellular processes that govern extracellular vesicle biology is essential to shed light on the physiological and pathological functions of these vesicles as well as on clinical applications involving their use and/or analysis. However, in this expanding field, much remains unknown regarding the origin, biogenesis, secretion, targeting and fate of these vesicles.
[16]
Pegtel D M, Gould S J. Exosomes. Annual Review of Biochemistry, 2019, 88: 487-514.
[17]
Battistelli M, Falcieri E. Apoptotic bodies: particular extracellular vesicles involved in intercellular communication. Biology, 2020, 9(1): 21.
In the last decade, a new method of cell–cell communication mediated by membranous extracellular vesicles (EVs) has emerged. EVs, including exosomes, microvesicles, and apoptotic bodies (ApoBDs), represent a new and important topic, because they are a means of communication between cells and they can also be involved in removing cellular contents. EVs are characterized by differences in size, origin, and content and different types have different functions. They appear as membranous sacs released by a variety of cells, in different physiological and patho-physiological conditions. Intringuingly, exosomes and microvesicles are a potent source of genetic information carriers between different cell types both within a species and even across a species barrier. New, and therefore still relatively poorly known vesicles are apoptotic bodies, on which numerous in-depth studies are needed in order to understand their role and possible function. In this review we would like to analyze their morpho-functional characteristics.
[18]
Sun H Y, Burrola S, Wu J C, et al. Extracellular vesicles in the development of cancer therapeutics. International Journal of Molecular Sciences, 2020, 21(17): 6097.
Extracellular vesicles (EVs) are small lipid bilayer-delimited nanoparticles released from all types of cells examined thus far. Several groups of EVs, including exosomes, microvesicles, and apoptotic bodies, have been identified according to their size and biogenesis. With extensive investigations on EVs over the last decade, it is now recognized that EVs play a pleiotropic role in various physiological processes as well as pathological conditions through mediating intercellular communication. Most notably, EVs have been shown to be involved in cancer initiation and progression and EV signaling in cancer are viewed as potential therapeutic targets. Furthermore, as membrane nanoparticles, EVs are natural products with some of them, such as tumor exosomes, possessing tumor homing propensity, thus leading to strategies utilizing EVs as drug carriers to effectively deliver cancer therapeutics. In this review, we summarize recent reports on exploring EVs signaling as potential therapeutic targets in cancer as well as on developing EVs as therapeutic delivery carriers for cancer therapy. Findings from preclinical studies are primarily discussed, with early phase clinical trials reviewed. We hope to provide readers updated information on the development of EVs as cancer therapeutic targets or therapeutic carriers.
[19]
Lo Cicero A, Stahl P D, Raposo G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Current Opinion in Cell Biology, 2015, 35: 69-77.
The release of extracellular vesicles (EVs) is a highly conserved process exploited by diverse organisms as a mode of intercellular communication. Vesicles of sizes ranging from 30 to 1000nm, or even larger, are generated by blebbing of the plasma membrane (microvesicles) or formed in multivesicular endosomes (MVEs) to be secreted by exocytosis as exosomes. Exosomes, microvesicles and other EVs contain membrane and cytosolic components that include proteins, lipids and RNAs, a composition that differs related to their site of biogenesis. Several mechanisms are involved in vesicle formation at the plasma membrane or in endosomes, which is reflected in their heterogeneity, size and composition. EVs have significant promise for therapeutics and diagnostics and for understanding physiological and pathological processes all of which have boosted research to find modulators of their composition, secretion and targeting. Copyright © 2015 Elsevier Ltd. All rights reserved.
[20]
Cheng Y W, Cao X, Qin L J. Mesenchymal stem cell-derived extracellular vesicles: a novel cell-free therapy for Sepsis. Frontiers in Immunology, 2020, 11: 647.
Sepsis remains a serious and life-threatening disease with high morbidity and mortality. Due to the unique immunomodulatory, anti-inflammatory, anti-apoptotic, anti-microbial, anti-oxidative, and reparative properties, mesenchymal stem cells (MSCs) have been extensively used in preclinical and clinical trials for diverse diseases and have shown great therapeutic potential in sepsis. However, concerns remain regarding whether MSCs can become tumorigenic or have other side effects. Extracellular vesicles (EVs) are a heterogeneous group of membrane-enclosed particles released from almost any cell and perform an important role in intercellular communication. Recently, it has emerged that EVs derived from MSCs (MSC-EVs) appear to exert a therapeutic benefit similar to MSCs in protecting against sepsis-induced organ dysfunction by delivering a cargo that includes RNAs and proteins to target cells. More importantly, compared to their parent cells, MSC-EVs have a superior safety profile, can be safely stored without losing function, and possess other advantages. Hence, MSC-EVs may be used as a novel alternative to MSC-based therapy in sepsis. Here, we summarize the properties and applications of MSC-EVs in sepsis.Copyright © 2020 Cheng, Cao and Qin.
[21]
Jafarinia M, Alsahebfosoul F, Salehi H, et al. Mesenchymal stem cell-derived extracellular vesicles: a novel cell-free therapy. Immunological Investigations, 2020, 49(7): 758-780.
[22]
Bartel S, Deshane J, Wilkinson T, et al. Extracellular vesicles as mediators of cellular cross talk in the lung microenvironment. Frontiers in Medicine, 2020, 7: 326.
The human lung is a complex tissue subdivided into several regions that differ in size, function, and resident cell types. Despite years of intensive research, we still do not fully understand the cross talk between these different regions and diverse cell populations in the lung and how this is altered in the development of chronic respiratory disease. The discovery of extracellular vesicles (EVs), small membrane vesicles released from cells for intercellular communication, has added another layer of complexity to cellular cross talk in the complex lung microenvironment. EVs from patients with chronic obstructive pulmonary disease, asthma, or sarcoidosis have been shown to carry microRNAs, proteins, and lipids that may contribute to inflammation or tissue degeneration. Here, we summarize the contribution of these small vesicles in the interplay of several different cell types in the lung microenvironment, with a focus on the development of chronic respiratory diseases. Although there are already many studies demonstrating the adverse effects of EVs in the diseased lung, we still have substantial knowledge gaps regarding the concrete role of EV involvement in lung disease, which should be addressed in future studies.Copyright © 2020 Bartel, Deshane, Wilkinson and Gabrielsson.
[23]
Doeppner T R, Herz J, Görgens A, et al. Extracellular vesicles improve post-stroke neuroregeneration and prevent postischemic immunosuppression. Stem Cells Translational Medicine, 2015, 4(10): 1131-1143.
Although the initial concepts of stem cell therapy aimed at replacing lost tissue, more recent evidence has suggested that stem and progenitor cells alike promote postischemic neurological recovery by secreted factors that restore the injured brain's capacity to reshape. Specifically, extracellular vesicles (EVs) derived from stem cells such as exosomes have recently been suggested to mediate restorative stem cell effects. In order to define whether EVs indeed improve postischemic neurological impairment and brain remodeling, we systematically compared the effects of mesenchymal stem cell (MSC)-derived EVs (MSC-EVs) with MSCs that were i.v. delivered to mice on days 1, 3, and 5 (MSC-EVs) or on day 1 (MSCs) after focal cerebral ischemia in C57BL6 mice. For as long as 28 days after stroke, motor coordination deficits, histological brain injury, immune responses in the peripheral blood and brain, and cerebral angiogenesis and neurogenesis were analyzed. Improved neurological impairment and long-term neuroprotection associated with enhanced angioneurogenesis were noticed in stroke mice receiving EVs from two different bone marrow-derived MSC lineages. MSC-EV administration closely resembled responses to MSCs and persisted throughout the observation period. Although cerebral immune cell infiltration was not affected by MSC-EVs, postischemic immunosuppression (i.e., B-cell, natural killer cell, and T-cell lymphopenia) was attenuated in the peripheral blood at 6 days after ischemia, providing an appropriate external milieu for successful brain remodeling. Because MSC-EVs have recently been shown to be apparently safe in humans, the present study provides clinically relevant evidence warranting rapid proof-of-concept studies in stroke patients.Transplantation of mesenchymal stem cells (MSCs) offers an interesting adjuvant approach next to thrombolysis for treatment of ischemic stroke. However, MSCs are not integrated into residing neural networks but act indirectly, inducing neuroprotection and promoting neuroregeneration. Although the mechanisms by which MSCs act are still elusive, recent evidence has suggested that extracellular vesicles (EVs) might be responsible for MSC-induced effects under physiological and pathological conditions. The present study has demonstrated that EVs are not inferior to MSCs in a rodent stroke model. EVs induce long-term neuroprotection, promote neuroregeneration and neurological recovery, and modulate peripheral post-stroke immune responses. Also, because EVs are well-tolerated in humans, as previously reported, the administration of EVs under clinical settings might set the path for a novel and innovative therapeutic stroke concept without the putative side effects attached to stem cell transplantation.©AlphaMed Press.
[24]
Kamerkar S, LeBleu V S, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 2017, 546(7659): 498-503.
[25]
Zhu X H, Badawi M, Pomeroy S, et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. Journal of Extracellular Vesicles, 2017, 6(1): 1324730.
[26]
Martin J D, Cabral H, Stylianopoulos T, et al. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nature Reviews Clinical Oncology, 2020, 17(4): 251-266.
Multiple nanotherapeutics have been approved for patients with cancer, but their effects on survival have been modest and, in some examples, less than those of other approved therapies. At the same time, the clinical successes achieved with immunotherapy have revolutionized the treatment of multiple advanced-stage malignancies. However, the majority of patients do not benefit from the currently available immunotherapies and many develop immune-related adverse events. By contrast, nanomedicines can reduce - but do not eliminate - the risk of certain life-threatening toxicities. Thus, the combination of these therapeutic classes is of intense research interest. The tumour microenvironment (TME) is a major cause of the failure of both nanomedicines and immunotherapies that not only limits delivery, but also can compromise efficacy, even when agents accumulate in the TME. Coincidentally, the same TME features that impair nanomedicine delivery can also cause immunosuppression. In this Perspective, we describe TME normalization strategies that have the potential to simultaneously promote the delivery of nanomedicines and reduce immunosuppression in the TME. Then, we discuss the potential of a combined nanomedicine-based TME normalization and immunotherapeutic strategy designed to overcome each step of the cancer-immunity cycle and propose a broadly applicable 'minimal combination' of therapies designed to increase the number of patients with cancer who are able to benefit from immunotherapy.
[27]
Davidson L M, Berkelhamer S K. Bronchopulmonary dysplasia: chronic lung disease of infancy and long-term pulmonary outcomes. Journal of Clinical Medicine, 2017, 6(1): 4.
[28]
Willis G R, Fernandez-Gonzalez A, Anastas J, et al. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. American Journal of Respiratory and Critical Care Medicine, 2018, 197(1): 104-116.
Mesenchymal stem/stromal cell (MSC) therapies have shown promise in preclinical models of pathologies relevant to newborn medicine, such as bronchopulmonary dysplasia (BPD). We have reported that the therapeutic capacity of MSCs is comprised in their secretome, and demonstrated that the therapeutic vectors are exosomes produced by MSCs (MSC-exos).To assess efficacy of MSC-exo treatment in a preclinical model of BPD and to investigate mechanisms underlying MSC-exo therapeutic action.Exosomes were isolated from media conditioned by human MSC cultures. Newborn mice were exposed to hyperoxia (HYRX; 75% O), treated with exosomes on Postnatal Day (PN) 4 and returned to room air on PN7. Treated animals and appropriate controls were harvested on PN7, -14, or -42 for assessment of pulmonary parameters.HYRX-exposed mice presented with pronounced alveolar simplification, fibrosis, and pulmonary vascular remodeling, which was effectively ameliorated by MSC-exo treatment. Pulmonary function tests and assessment of pulmonary hypertension showed functional improvements after MSC-exo treatment. Lung mRNA sequencing demonstrated that MSC-exo treatment induced pleiotropic effects on gene expression associated with HYRX-induced inflammation and immune responses. MSC-exos modulate the macrophage phenotype fulcrum, suppressing the proinflammatory "M1" state and augmenting an antiinflammatory "M2-like" state, both in vitro and in vivo.MSC-exo treatment blunts HYRX-associated inflammation and alters the hyperoxic lung transcriptome. This results in alleviation of HYRX-induced BPD, improvement of lung function, decrease in fibrosis and pulmonary vascular remodeling, and amelioration of pulmonary hypertension. The MSC-exo mechanism of action is associated with modulation of lung macrophage phenotype.
[29]
Chaubey S, Thueson S, Ponnalagu D, et al. Early gestational mesenchymal stem cell secretome attenuates experimental bronchopulmonary dysplasia in part via exosome-associated factor TSG-6. Stem Cell Research & Therapy, 2018, 9(1): 173.
[30]
Abele A N, Taglauer E S, Almeda M, et al. Antenatal mesenchymal stromal cell extracellular vesicle treatment preserves lung development in a model of bronchopulmonary dysplasia due to chorioamnionitis. American Journal of Physiology. Lung Cellular and Molecular Physiology, 2022, 322(2): L179-L190.
[31]
Ahn S Y, Park W S, Kim Y E, et al. Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Experimental & Molecular Medicine, 2018, 50(4): 1-12.
[32]
Lopes-Pacheco M, Robba C, Rocco P, et al. Current understanding of the therapeutic benefits of mesenchymal stem cells in acute respiratory distress syndrome. Cell Biol Toxicol, 2019, 36: 83.
[33]
Abraham A, Krasnodembskaya A. Mesenchymal stem cell-derived extracellular vesicles for the treatment of acute respiratory distress syndrome. Stem Cells Translational Medicine, 2020, 9(1): 28-38.
Acute respiratory distress syndrome (ARDS) is a serious and potentially fatal acute inflammatory lung condition which currently has no specific treatments targeting its pathophysiology. However, mesenchymal stem cells have been shown to have very promising therapeutic potential, and recently, it has been established that their effect is largely due to the transfer of extracellular vesicles (EVs). EVs have been shown to transfer a variety of substances such as mRNA, miRNA, and even organelles such as mitochondria in order to ameliorate ARDS in preclinical models. In addition, the fact that they have been proven to have the same effect as their parent cells combined with their numerous advantages over whole cell administration means that they are a promising candidate for clinical application that merits further research.© 2019 The Authors. STEM CELLS TRANSLATIONAL MEDICINE published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.
[34]
Khatri M, Richardson L A, Meulia T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Research & Therapy, 2018, 9(1): 17.
[35]
Hu S, Park J, Liu A, et al. Mesenchymal stem cell microvesicles restore protein permeability across primary cultures of injured human lung microvascular endothelial cells. Stem Cells Translational Medicine, 2018, 7(8): 615-624.
Our previous study demonstrated that mesenchymal stem cell (MSC) microvesicles (MV) reduced lung inflammation, protein permeability, and pulmonary edema in endotoxin-induced acute lung injury in mice. However, the underlying mechanisms for restoring lung protein permeability were not fully understood. In this current study, we hypothesized that MSC MV would restore protein permeability across injured human lung microvascular endothelial cells (HLMVEC) in part through the transfer of angiopoietin-1 (Ang1) mRNA to the injured endothelium. A transwell coculture system was used to study the effect of MSC MV on protein permeability across HLMVECs injured by cytomix, a mixture of IL-1β, TNF-α, and IFN-γ (50 ng/ml). Our result showed that cytomix significantly increased permeability to FITC-dextran (70 kDa) across HLMVECs over 24 hours. Administration of MSC MVs restored this permeability in a dose dependent manner, which was associated with an increase in Ang1 mRNA and protein secretion in the injured endothelium. This beneficial effect was diminished when MSC MV was pretreated with an anti-CD44 antibody, suggesting that internalization of MV into the HLMVEC was required for the therapeutic effect. Fluorescent microscopy showed that MSC MV largely prevented the reorganization of cytoskeleton protein F-actin into "actin stress fiber" and restored the location of the tight junction protein ZO-1 and adherens junction protein VE-cadherin in injured HLMVECs. Ang1 siRNA pretreatment of MSC MV prior to administration to injured HLMVECs eliminated the therapeutic effect of MV. In summary, MSC MVs restored protein permeability across HLMVEC in part by increasing Ang1 secretion by injured HLMVEC. Stem Cells Translational Medicine 2018;7:615-624.© 2018 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.
[36]
Li J W, Wei L, Han Z, et al. Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21-5p. European Journal of Pharmacology, 2019, 852: 68-76.
MiR-21-5p is an anti-apoptotic miRNA known to mediate the protective effect of mesenchymal stromal cell-secreted exosomes (MSC-Exo) against oxidative stress-induced cell death. In the present research we employed murine lung ischemia/reperfusion (I/R) model and in vitro hypoxia/reoxygenation (H/R) model using primary murine pulmonary endothelial cells to investigate whether MSC-Exo could alleviate lung IRI by transporting miR-21-5p. Our data suggested that intratracheal administration of MSC-Exo or miR-21-5p agomir significantly reduced lung edema and dysfunction, M1 polarization of alveolar macrophages as well as secretion of HMGB1, IL-8, IL-1β, IL-6, IL-17 and TNF-α. Pre-challenge of MSCs by H/R significant increased miR-21-5p expression level in exosomes they secreted and the anti-IRI effect of these MSC-Exo, while pre-treatment of MSCs with miR-21-5p antagomir showed opposite effect. We further demonstrated that MSC-Exo ameliorated IRI in vivo or H/R induced apoptosis in vitro by inhibiting both intrinsic and extrinsic apoptosis pathway via miR-21-5p targeting PTEN and PDCD4, while artificial overexpressing PTEN or PDCD4 significantly attenuated the anti-apoptotic effect of MSC-Exo in vitro. Treatment with miR-21-5p agomir mimicked the IRI-reducing and anti-apoptotic effect of MSC-Exo. Our data suggested that MSC-Exo alleviate IRI in lung in an exosomal miR-21-5p-dependent manner. Treatment with MSC-Exo or miR-21-5p agomir might ameliorate IRI in lung.Copyright © 2019 Elsevier B.V. All rights reserved.
[37]
Yi X, Wei X, Lv H, et al. Exosomes derived from microRNA-30b-3p-overexpressing mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting SAA3. Experimental Cell Research, 2019, 383(2): 111454.
[38]
Chen W X, Zhou J, Zhou S S, et al. Microvesicles derived from human Wharton’s jelly mesenchymal stem cells enhance autophagy and ameliorate acute lung injury via delivery of miR-100. Stem Cell Research & Therapy, 2020, 11(1): 113.
[39]
Hao Q, Gudapati V, Monsel A, et al. Mesenchymal stem cell-derived extracellular vesicles decrease lung injury in mice. Journal of Immunology (Baltimore, Md. : 1950), 2019, 203(7): 1961-1972.
Human mesenchymal stem cell (MSC) extracellular vesicles (EV) can reduce the severity of bacterial pneumonia, but little is known about the mechanisms underlying their antimicrobial activity. In the current study, we found that bacterial clearance induced by MSC EV in Escherichia coli pneumonia in C57BL/6 mice was associated with high levels of leukotriene (LT) B4 in the injured alveolus. More importantly, the antimicrobial effect of MSC EV was abrogated by cotreatment with a LTB4 BLT1 antagonist. To determine the role of MSC EV on LT metabolism, we measured the effect of MSC EV on a known ATP-binding cassette transporter, multidrug resistance–associated protein 1 (MRP1), and found that MSC EV suppressed MRP1 mRNA, protein, and pump function in LPS-stimulated Raw264.7 cells in vitro. The synthesis of LTB4 and LTC4 from LTA4 are competitive, and MRP1 is the efflux pump for LTC4. Inhibition of MRP1 will increase LTB4 production. In addition, administration of a nonspecific MRP1 inhibitor (MK-571) reduced LTC4 and subsequently increased LTB4 levels in C57BL/6 mice with acute lung injury, increasing overall antimicrobial activity. We previously found that the biological effects of MSC EV were through the transfer of its content, such as mRNA, microRNA, and proteins, to target cells. In the current study, miR-145 knockdown abolished the effect of MSC EV on the inhibition of MRP1 in vitro and the antimicrobial effect in vivo. In summary, MSC EV suppressed MRP1 activity through transfer of miR-145, thereby resulting in enhanced LTB4 production and antimicrobial activity through LTB4/BLT1 signaling.
[40]
Song Y, Dou H, Li X, et al. Exosomal miR-146a contributes to the enhanced therapeutic efficacy of interleukin-1β-primed mesenchymal stem cells against sepsis. Stem Cells (Dayton, Ohio), 2017, 35(5): 1208-1221.
Improving the immunomodulatory efficacy of mesenchymal stem cells (MSCs) through pretreatment with pro-inflammatory cytokines is an evolving field of investigation. However, the underlying mechanisms have not been fully clarified. Here, we pretreated human umbilical cord-derived MSCs with interleukin-1β (IL-1β) and evaluated their therapeutic effects in a cecal ligation and puncture-induced sepsis model. We found that systemic administration of IL-1β-pretreated MSCs (βMSCs) ameliorated the symptoms of murine sepsis more effectively and increased the survival rate compared with naïve MSCs. Furthermore, βMSCs could more effectively induce macrophage polarization toward an anti-inflammatory M2 phenotype through the paracrine activity. Mechanistically, we demonstrated that βMSC-derived exosomes contributed to the enhanced immunomodulatory properties of βMSCs both in vitro and in vivo. Importantly, we found that miR-146a, a well-known anti-inflammatory microRNA, was strongly upregulated by IL-1β stimulation and selectively packaged into exosomes. This exosomal miR-146a was transferred to macrophages, resulted in M2 polarization, and finally led to increased survival in septic mice. In contrast, inhibition of miR-146a through transfection with miR-146a inhibitors partially negated the immunomodulatory properties of βMSC-derived exosomes. Taken together, IL-1β pretreatment effectively enhanced the immunomodulatory properties of MSCs partially through exosome-mediated transfer of miR-146a. Therefore, we believe that IL-1β pretreatment may provide a new modality for better therapeutic application of MSCs in inflammatory disorders.
[41]
Wang J, Huang R, Xu Q, et al. Mesenchymal stem cell-derived extracellular vesicles alleviate acute lung injury via transfer of miR-27a-3p. Critical Care Medicine, 2020, 48(7): e599-e610.
[42]
Morrison T J, Jackson M V, Cunningham E K, et al. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. American Journal of Respiratory and Critical Care Medicine, 2017, 196(10): 1275-1286.
Acute respiratory distress syndrome (ARDS) remains a major cause of respiratory failure in critically ill patients. Mesenchymal stromal cells (MSCs) are a promising candidate for a cell-based therapy. However, the mechanisms of MSCs' effects in ARDS are not well understood. In this study, we focused on the paracrine effect of MSCs on macrophage polarization and the role of extracellular vesicle (EV)-mediated mitochondrial transfer.To determine the effects of human MSCs on macrophage function in the ARDS environment and to elucidate the mechanisms of these effects.Human monocyte-derived macrophages (MDMs) were studied in noncontact coculture with human MSCs when stimulated with LPS or bronchoalveolar lavage fluid (BALF) from patients with ARDS. Murine alveolar macrophages (AMs) were cultured ex vivo with/without human MSC-derived EVs before adoptive transfer to LPS-injured mice.MSCs suppressed cytokine production, increased M2 macrophage marker expression, and augmented phagocytic capacity of human MDMs stimulated with LPS or ARDS BALF. These effects were partially mediated by CD44-expressing EVs. Adoptive transfer of AMs pretreated with MSC-derived EVs reduced inflammation and lung injury in LPS-injured mice. Inhibition of oxidative phosphorylation in MDMs prevented the modulatory effects of MSCs. Generating dysfunctional mitochondria in MSCs using rhodamine 6G pretreatment also abrogated these effects.In the ARDS environment, MSCs promote an antiinflammatory and highly phagocytic macrophage phenotype through EV-mediated mitochondrial transfer. MSC-induced changes in macrophage phenotype critically depend on enhancement of macrophage oxidative phosphorylation. AMs treated with MSC-derived EVs ameliorate lung injury in vivo.
[43]
Moss B J, Ryter S W, Rosas I O. Pathogenic mechanisms underlying idiopathic pulmonary fibrosis. Annual Review of Pathology, 2022, 17: 515-546.
[44]
Schäfer S, Funke-Chambour M, Berezowska S. Idiopathic pulmonary fibrosis-epidemiology, causes, and clinical course. Der Pathologe, 2020, 41(1): 46-51.
[45]
Mansouri N, Willis G R, Fernandez-Gonzalez A, et al. Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes. JCI Insight, 2019, 4(21): e128060.
[46]
Xu C, Zhao J, Li Q, et al. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Research & Therapy, 2020, 11(1): 503.
[47]
Zhang E, Geng X, Shan S, et al. Exosomes derived from bone marrow mesenchymal stem cells reverse epithelial-mesenchymal transition potentially via attenuating Wnt/β-catenin signaling to alleviate silica-induced pulmonary fibrosis. Toxicology Mechanisms and Methods, 2021, 31(9): 655-666.
[48]
Sun L, Zhu M, Feng W, et al. Exosomal miRNA Let-7 from menstrual blood-derived endometrial stem cells alleviates pulmonary fibrosis through regulating mitochondrial DNA damage. Oxidative Medicine and Cellular Longevity, 2019, 2019: 4506303.
[49]
Gao Y, Sun J, Dong C, et al. Extracellular vesicles derived from adipose mesenchymal stem cells alleviate PM2.5-induced lung injury and pulmonary fibrosis. Medical Science Monitor : International Medical Journal of Experimental and Clinical Research, 2020, 26: e922782.
[50]
Wan X, Chen S, Fang Y, et al. Mesenchymal stem cell-derived extracellular vesicles suppress the fibroblast proliferation by downregulating FZD 6 expression in fibroblasts via micrRNA-29b-3p in idiopathic pulmonary fibrosis. Journal of Cellular Physiology, 2020, 235(11): 8613-8625.
[51]
Zhou J, Lin Y, Kang X, et al. microRNA-186 in extracellular vesicles from bone marrow mesenchymal stem cells alleviates idiopathic pulmonary fibrosis via interaction with SOX4 and DKK1. Stem Cell Research & Therapy, 2021, 12(1): 96.
[52]
Villamizar O, Waters S A, Scott T, et al. Mesenchymal stem cell exosome delivered zinc finger protein activation of cystic fibrosis transmembrane conductance regulator. Journal of Extracellular Vesicles, 2021, 10(3): e12053.
[53]
Agustí A, Hogg J C. Update on the pathogenesis of chronic obstructive pulmonary disease. The New England Journal of Medicine, 2019, 381(13): 1248-1256.
[54]
Rovina N, Koutsoukou A, Koulouris N G. Inflammation and immune response in COPD: where do we stand? Mediators of Inflammation, 2013, 2013: 413735.
[55]
Harrell C R, Miloradovic D, Sadikot R, et al. Molecular and cellular mechanisms responsible for beneficial effects of mesenchymal stem cell-derived product “Exo-d-MAPPS” in attenuation of chronic airway inflammation. Analytical Cellular Pathology (Amsterdam), 2020, 2020: 3153891.
[56]
Kim Y S, Kim J Y, Cho R, et al. Adipose stem cell-derived nanovesicles inhibit emphysema primarily via an FGF2-dependent pathway. Experimental & Molecular Medicine, 2017, 49(1): e284.
[57]
Maremanda k P, Sundar I K, Rahman I. Protective role of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice. Toxicology and Applied Pharmacology, 2019, 385: 114788.
[58]
Hoy R F, Chambers D C. Silica-related diseases in the modern world. Allergy, 2020, 75(11): 2805-2817.
[59]
Li X, An G, Wang Y, et al. Targeted migration of bone marrow mesenchymal stem cells inhibits silica-induced pulmonary fibrosis in rats. Stem Cell Research & Therapy, 2018, 9(1): 335.
[60]
Choi M, Ban T, Rhim T. Therapeutic use of stem cell transplantation for cell replacement or cytoprotective effect of microvesicle released from mesenchymal stem cell. Molecules and Cells, 2014, 37(2): 133-139.
Idiopathic pulmonary fibrosis (IPF) is the most common and severe type of idiopathic interstitial pneumonias (IIP), and which is currently no method was developed to restore normal structure and function. There are several reports on therapeutic effects of adult stem cell transplantations in animal models of pulmonary fibrosis. However, little is known about how mesenchymal stem cell (MSC) can repair the IPF. In this study, we try to provide the evidence to show that transplanted mesenchymal stem cells directly replace fibrosis with normal lung cells using IPF model mice. As results, transplanted MSC successfully integrated and differentiated into type II lung cell which express surfactant protein. In the other hand, we examine the therapeutic effects of microvesicle treatment, which were released from mesenchymal stem cells. Though the therapeutic effects of MV treatment is less than that of MSC treatment, MV treatment meaningfully reduced the symptom of IPF, such as collagen deposition and inflammation. These data suggest that stem cell transplantation may be an effective strategy for the treatment of pulmonary fibrosis via replacement and cytoprotective effect of microvesicle released from MSCs.
[61]
Phinney D G, Di Giuseppe M, Njah J, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nature Communications, 2015, 6: 8472.
Phinney, Donald G.; Boregowda, Siddaraju V. Scripps Res Inst, Dept Mol Therapeut, Jupiter, FL 33458 USA. Di Giuseppe, Michelangelo; Njah, Joel; St Croix, Claudette M.; Di, Y. Peter; Leikauf, George D.; Ortiz, Luis A. Univ Pittsburgh, Dept Environm & Occupat Hlth, Pittsburgh, PA 15219 USA. Sala, Ernest Hosp Son Espases, Palma De Mallorca 07010, Spain. Shiva, Sruti; St Croix, Claudette M. Univ Pittsburgh, Dept Pharmacol, Pittsburgh, PA 15219 USA. St Croix, Claudette M.; Stolz, Donna B.; Watkins, Simon C. Univ Pittsburgh, Dept Cell Biol, Pittsburgh, PA 15219 USA. Kolls, Jay Univ Pittsburgh, Mellon Fdn, Pediat Res Inst, Pittsburgh, PA 15219 USA. Riches, David W. H. Natl Jewish Hlth, Dept Pediat, Denver, CO 80206 USA. Deiuliis, Giuseppe; Kaminski, Naftali Yale Univ, Dept Med, New Haven, CT 06510 USA. McKenna, David H. Univ Minnesota, Dept Lab Med & Pathol, St Paul, MN 55108 USA.
[62]
Bandeira E, Oliveira H, Silva J D, et al. Therapeutic effects of adipose-tissue-derived mesenchymal stromal cells and their extracellular vesicles in experimental silicosis. Respiratory Research, 2018, 19(1): 104.
Background: Silicosis is an occupational disease that affects workers who inhale silica particles, leading to extensive lung fibrosis and ultimately causing respiratory failure. Mesenchymal stromal cells (MSCs) have been shown to exert therapeutic effects in lung diseases and represent an alternative treatment for silicosis. Recently, it has been suggested that similar effects can be achieved by the therapeutic use of extracellular vesicles (EVs) obtained from MSCs. The aim of this study was to investigate the effects of adipose-tissue-derived MSCs (AD-MSCs) or their EVs in a model of silicosis.Methods: Silicosis was induced by intratracheal instillation of silica in C57BL/6 mice. After the onset of disease, animals received saline, AD-MSCs, or EVs, intratracheally.Results: At day 30, AD-MSCs and EVs led to a reduction in collagen fiber content, size of granuloma, and in the number of macrophages inside granuloma and in the alveolar septa. In addition, the expression levels of interleukin 1 beta and transforming growth factor beta in the lungs were decreased. Higher dose of EVs also reduced lung static elastance when compared with the untreated silicosis group.Conclusions: Both AD-MSCs and EVs, locally delivered, ameliorated fibrosis and inflammation, but dose-enhanced EVs yielded better therapeutic outcomes in this model of silicosis.
[63]
Papi A, Brightling C, Pedersen S E, et al. Asthma. Lancet (London, England), 2018, 391(10122): 783-800.
[64]
Boldrini-Leite L M, Michelotto P V Jr, de Moura S A B, et al. Lung tissue damage associated with allergic asthma in BALB/c mice could be controlled with a single injection of mesenchymal stem cells from human bone marrow up to 14 d after transplantation. Cell Transplantation, 2020, 29: 963689720913254.
[65]
Du Y M, Zhuansun Y X, Chen R, et al. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Experimental Cell Research, 2018, 363(1): 114-120.
[66]
de Castro L L, Xisto D G, Kitoko J Z, et al. Human adipose tissue mesenchymal stromal cells and their extracellular vesicles act differentially on lung mechanics and inflammation in experimental allergic asthma. Stem Cell Research & Therapy, 2017, 8(1): 151.
[67]
Fang S B, Zhang H Y, Wang C, et al. Small extracellular vesicles derived from human mesenchymal stromal cells prevent group 2 innate lymphoid cell-dominant allergic airway inflammation through delivery of miR-146a-5p. Journal of Extracellular Vesicles, 2020, 9(1): 1723260.
[68]
Dong L Y, Wang Y, Zheng T T, et al. Hypoxic hUCMSC-derived extracellular vesicles attenuate allergic airway inflammation and airway remodeling in chronic asthma mice. Stem Cell Research & Therapy, 2021, 12(1): 4.
[69]
Guazzi M, Naeije R. Pulmonary hypertension in heart failure: pathophysiology, pathobiology, and emerging clinical perspectives. Journal of the American College of Cardiology, 2017, 69(13): 1718-1734.
Pulmonary hypertension is a common hemodynamic complication of heart failure. Interest in left-sided pulmonary hypertension has increased remarkably in recent years because its development and consequences for the right heart are now seen as mainstay abnormalities that begin in the early stages of the disease and bear unfavorable prognostic insights. However, some knowledge gaps limit our ability to influence this complex condition. Accordingly, attention is now focused on: 1) establishing a definitive consensus for a hemodynamic definition, perhaps incorporating exercise and fluid challenge; 2) implementing the limited data available on the pathobiology of lung capillaries and small arteries; 3) developing standard methods for assessing right ventricular function and, hopefully, its coupling to pulmonary circulation; and 4) searching for effective therapies that may benefit lung vessels and the remodeled right ventricle. The authors review the pathophysiology, pathobiology, and emerging clinical perspectives on pulmonary hypertension across the broad spectrum of heart failure stages.Copyright © 2017 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.
[70]
Lee C J, Alex Mitsialis S, Aslam M, et al. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation, 2012, 126(22): 2601-2611.
Hypoxia induces an inflammatory response in the lung manifested by alternative activation of macrophages with elevation of proinflammatory mediators that are critical for the later development of hypoxic pulmonary hypertension. Mesenchymal stromal cell transplantation inhibits lung inflammation, vascular remodeling, and right heart failure and reverses hypoxic pulmonary hypertension in experimental models of disease. In this study, we aimed to investigate the paracrine mechanisms by which mesenchymal stromal cells are protective in hypoxic pulmonary hypertension.We fractionated mouse mesenchymal stromal cell-conditioned media to identify the biologically active component affecting in vivo hypoxic signaling and determined that exosomes, secreted membrane microvesicles, suppressed the hypoxic pulmonary influx of macrophages and the induction of proinflammatory and proproliferative mediators, including monocyte chemoattractant protein-1 and hypoxia-inducible mitogenic factor, in the murine model of hypoxic pulmonary hypertension. Intravenous delivery of mesenchymal stromal cell-derived exosomes (MEX) inhibited vascular remodeling and hypoxic pulmonary hypertension, whereas MEX-depleted media or fibroblast-derived exosomes had no effect. MEX suppressed the hypoxic activation of signal transducer and activator of transcription 3 (STAT3) and the upregulation of the miR-17 superfamily of microRNA clusters, whereas it increased lung levels of miR-204, a key microRNA, the expression of which is decreased in human pulmonary hypertension. MEX produced by human umbilical cord mesenchymal stromal cells inhibited STAT3 signaling in isolated human pulmonary artery endothelial cells, demonstrating a direct effect of MEX on hypoxic vascular cells.This study indicates that MEX exert a pleiotropic protective effect on the lung and inhibit pulmonary hypertension through suppression of hyperproliferative pathways, including STAT3-mediated signaling induced by hypoxia.
[71]
Chen J Y, An R, Liu Z J, et al. Therapeutic effects of mesenchymal stem cell-derived microvesicles on pulmonary arterial hypertension in rats. Acta Pharmacologica Sinica, 2014, 35(9): 1121-1128.
[72]
Aliotta J M, Pereira M, Wen S C, et al. Exosomes induce and reverse monocrotaline-induced pulmonary hypertension in mice. Cardiovascular Research, 2016, 110(3): 319-330.
Extracellular vesicles (EVs) from mice with monocrotaline (MCT)-induced pulmonary hypertension (PH) induce PH in healthy mice, and the exosomes (EXO) fraction of EVs from mesenchymal stem cells (MSCs) can blunt the development of hypoxic PH. We sought to determine whether the EXO fraction of EVs is responsible for modulating pulmonary vascular responses and whether differences in EXO-miR content explains the differential effects of EXOs from MSCs and mice with MCT-PH.Plasma, lung EVs from MCT-PH, and control mice were divided into EXO (exosome), microvesicle (MV) fractions and injected into healthy mice. EVs from MSCs were divided into EXO, MV fractions and injected into MCT-treated mice. PH was assessed by right ventricle-to-left ventricle + septum (RV/LV + S) ratio and pulmonary arterial wall thickness-to-diameter (WT/D) ratio. miR microarray analyses were also performed on all EXO populations. EXOs but not MVs from MCT-injured mice increased RV/LV + S, WT/D ratios in healthy mice. MSC-EXOs prevented any increase in RV/LV + S, WT/D ratios when given at the time of MCT injection and reversed the increase in these ratios when given after MCT administration. EXOs from MCT-injured mice and patients with idiopathic pulmonary arterial hypertension (IPAH) contained increased levels of miRs-19b,-20a,-20b, and -145, whereas miRs isolated from MSC-EXOs had increased levels of anti-inflammatory, anti-proliferative miRs including miRs-34a,-122,-124, and -127.These findings suggest that circulating or MSC-EXOs may modulate pulmonary hypertensive effects based on their miR cargo. The ability of MSC-EXOs to reverse MCT-PH offers a promising potential target for new PAH therapies.Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2016. For permissions please email: journals.permissions@oup.com.
[73]
Zhang C, Wang P, Mohammed A, et al. Function of adipose-derived mesenchymal stem cells in monocrotaline-induced pulmonary arterial hypertension through miR-191 via regulation of BMPR2. BioMed Research International, 2019, 2019: 2858750.
[74]
Hogan S E, Rodriguez Salazar M P, Cheadle J, et al. Mesenchymal stromal cell-derived exosomes improve mitochondrial health in pulmonary arterial hypertension. American Journal of Physiology. Lung Cellular and Molecular Physiology, 2019, 316(5): L723-L737.
PDF(663 KB)

1198

Accesses

0

Citation

Detail

Sections
Recommended

/