HUC-MSCs

hUC - MSCs
  • 文章类型: Journal Article
    由中风引起的脑损伤具有很高的死亡率,并且仍然是世界范围内的主要医学挑战。近年来,人脐带间充质干细胞(hUC-MSCs)在不同的成人和新生儿卒中动物模型中治疗卒中的应用受到了极大的关注.然而,使用hUC-MSCs通过全身给药治疗缺血性卒中尚未得到充分研究。在这项研究中,我们进行了多种实验来探讨hUC-MSCs对大鼠的神经保护作用。我们的发现表明,以2×10^7细胞/kg的高剂量静脉注射hUC-MSCs可显着改善缺血性中风引起的脑损伤。在诱导大鼠短暂性大脑中动脉阻塞(MCAO)和随后的再灌注后一天,观察到了这种改善。值得注意的是,这种单次给予hUC-MSCs的疗效超过了依达拉奉,即使后者连续使用三天。机械上,来自hUC-MSCs的分泌因子,如HGF,BDNF,和TNFR1,改善MDA和T-SOD水平以调节氧化应激。特别是,TNFR1还改善了与氧化应激相关的重要蛋白NQO-1和HO-1的表达。更重要的是,TNFR1通过调节血液中的IL-6水平在减少炎症中起重要作用。此外,观察到TNFR1影响血脑屏障(BBB)的通透性,如伊万蓝实验和ZO-1的蛋白质表达所示。这项研究代表了传统方法的突破,并为临床药物和试验提供了新的策略。
    Brain injury caused by stroke has a high rate of mortality and remains a major medical challenge worldwide. In recent years, there has been significant attention given to the use of human Umbilical cord-derived Mesenchymal Stem Cells (hUC-MSCs) for the treatment of stroke in different adult and neonate animal models of stroke. However, using hUC-MSCs by systemic administration to treat ischemic stroke has not been investigated sufficiently. In this study, we conducted various experiments to explore the neuroprotection of hUC-MSCs in rats. Our findings demonstrate that an intravenous injection of a high dose of hUC-MSCs at 2 × 10^7 cells/kg markedly ameliorated brain injury resulting from ischemic stroke. This improvement was observed one day after inducing transient middle cerebral artery occlusion (MCAO) and subsequent reperfusion in rats. Notably, the efficacy of this single administration of hUC-MSCs surpassed that of edaravone, even when the latter was used continuously over three days. Mechanistically, secretory factors derived from hUC-MSCs, such as HGF, BDNF, and TNFR1, ameliorated the levels of MDA and T-SOD to regulate oxidative stress. In particular, TNFR1 also improved the expression of NQO-1 and HO-1, important proteins associated with oxidative stress. More importantly, TNFR1 played a significant role in reducing inflammation by modulating IL-6 levels in the blood. Furthermore, TNFR1 was observed to influence the permeability of the blood-brain barrier (BBB) as demonstrated in the evan\'s blue experiment and protein expression of ZO-1. This study represented a breakthrough in traditional methods and provided a novel strategy for clinical medication and trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    神经性疼痛已被认为是最严重的慢性疼痛亚型之一,并对患者的身体和精神造成无法忍受的痛苦。本研究旨在验证人脐带间充质干细胞(HUC-MSCs)对慢性缩窄性损伤(CCI)诱导的神经病理性疼痛大鼠的镇痛作用及其通过调节小胶质细胞的作用机制。
    30只雄性SD大鼠随机分为三组(每组10只):假盐水组(S&S组),CCI+盐水组(C&S组)和CCI+HUC-MSCs组(C&U组)。在建模后的第7天,通过尾静脉注射盐水或HUC-MSC。在建模之前(第0天)和之后(第1、3、5、7、9、11、13和15天)测量结扎侧的爪机械缩回阈值(PMWT)和热缩回潜伏期(TWL)。建模后的第15天,免疫印迹和免疫荧光染色用于评估Iba-1(活化小胶质细胞的典型生物标志物)在脊髓背角结扎侧的表达丰度,透射电镜观察坐骨神经结扎的超微结构变化。
    与S&S组相比,C&S组的PMWT和TWL在第5天显著下降,然后持续到建模后第15天(C&SvsS&S,P<0.05),而在C&U组中观察到机械性痛觉过敏(第13天、第15天)和热异常性疼痛(第9天、第11天、第15天)的显著改善(C&UvsC&S,P<0.05)。同时,根据蛋白质印迹和免疫荧光染色分析,C&U组系统输注HUC-MSCs可显著抑制Iba-1的表达(P<0.05)。借助TEM检测,我们直观地注意到坐骨神经结扎的层状结构的有效重建,消除线粒体肿胀,在C&U组建模后第15天发现新的髓鞘形成。
    总的来说,静脉内给予HUC-MSCs通过抑制受损脊髓背角的小胶质细胞活化和减轻坐骨神经损伤,对CCI诱导的SD大鼠神经病理性疼痛有系统的改善作用。我们的发现为进一步开发基于HUC-MSCs的用于神经性疼痛的细胞疗法提供了新的参考。
    UNASSIGNED: Neuropathic pain has been considered as one of the most serious chronic pain subtypes and causes intolerable suffering to patients physically and mentally. This study aimed to verify the analgesic effect of intravenous administration of human umbilical cord mesenchymal stem cells (HUC-MSCs) upon rats with chronic constriction injury (CCI)-induced neuropathic pain and the concomitant mechanism via modulating microglia.
    UNASSIGNED: 30 male SD rats were randomized divided into three groups (n = 10 per group): Sham + Saline group (S&S group), CCI + Saline group (C&S group) and CCI + HUC-MSCs group (C&U group). Rats were injected with either saline or HUC-MSCs via the caudal vein on the 7th day after modelling. The paw mechanical withdrawal threshold (PMWT) and thermal withdrawal latency (TWL) of the ligation side were measured before (day 0) and after (day 1, 3, 5, 7, 9, 11, 13, and 15) modelling. On day 15 after modelling, western-blotting and immunofluorescent staining were used to assess the expressive abundance of Iba-1 (a typical biomarker of activated microglia) in the ligation side of the spinal cord dorsal horn, and ultrastructural changes of the ligation of sciatic nerve were evaluated by transmission electron microscope (TEM).
    UNASSIGNED: Compared with the S&S group, PMWT and TWL in the C&S group were significantly decreased on day 5 and then persisted to day 15 after modelling (C&S vs S&S, P < 0.05), while a significant amelioration of mechanical hyperalgesia (day 13, day 15) and thermal allodynia (day 9, day 11, day 15) was observed in the C&U group (C&U vs C&S, P < 0.05). Meanwhile, the expression of Iba-1 was significantly suppressed by systemic infusion of HUC-MSCs in the C&U group according to western-blotting and immunofluorescent staining analyses (P < 0.05). With the aid of TEM detection, we intuitively noticed the efficacious reconstruction of the laminate structure of the sciatic nerve ligation, elimination of mitochondrial swelling, and formation of new myelination were noted on day 15 after modelling in the C&U group.
    UNASSIGNED: Overall, intravenous administration of HUC-MSCs systemically revealed an ameliorative effect upon CCI-induced neuropathic pain in SD rats by inhibiting microglia activation in the dorsal horn of the impaired spinal cord and alleviating sciatic nerve injury. Our findings supply new references for the further development of HUC-MSCs-based cytotherapy for neuropathic pain administration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脊髓损伤(SCI)是一种导致运动和感觉功能受损的创伤性疾病。铁性凋亡是脊髓神经细胞死亡和神经功能丧失的主要原因之一,和铁凋亡抑制剂可有效减轻炎症和修复SCI。人脐带间充质干细胞(Huc-MSCs)虽然可以改善SCI患者的炎症微环境,促进神经再生,但其疗效受到SCI后局部微环境的限制。因此,在这项研究中,我们构建了一个协同Huc-MSCs和铁凋亡抑制剂的药物释放纳米粒系统,其中我们通过基于CD44靶向序列的Tz-A6肽锚定Huc-MSCs,并在另一端与活性氧(ROS)响应性药物纳米载体mPEG-b-Lys-BECI-TCO结合用于SCI修复。同时,我们还修改了经典的铁凋亡抑制剂Ferrostatin-1(Fer-1),并合成了一种新的前药Feborastatin-1(Feb-1)。结果表明,该治疗方案能显著抑制SCI后大鼠的铁蛋白凋亡和炎症反应,促进SCI大鼠神经功能恢复。本研究开发了一种治疗SCI的联合疗法,也为药物协同细胞治疗系统的构建提供了新的策略。
    Spinal cord injury (SCI) is a traumatic condition that results in impaired motor and sensory function. Ferroptosis is one of the main causes of neural cell death and loss of neurological function in the spinal cord, and ferroptosis inhibitors are effective in reducing inflammation and repairing SCI. Although human umbilical cord mesenchymal stem cells (Huc-MSCs) can ameliorate inflammatory microenvironments and promote neural regeneration in SCI, their efficacy is greatly limited by the local microenvironment after SCI. Therefore, in this study, we constructed a drug-release nanoparticle system with synergistic Huc-MSCs and ferroptosis inhibitor, in which we anchored Huc-MSCs by a Tz-A6 peptide based on the CD44-targeting sequence, and combined with the reactive oxygen species (ROS)-responsive drug nanocarrier mPEG-b-Lys-BECI-TCO at the other end for SCI repair. Meanwhile, we also modified the classic ferroptosis inhibitor Ferrostatin-1 (Fer-1) and synthesized a new prodrug Feborastatin-1 (Feb-1). The results showed that this treatment regimen significantly inhibited the ferroptosis and inflammatory response after SCI, and promoted the recovery of neurological function in rats with SCI. This study developed a combination therapy for the treatment of SCI and also provides a new strategy for the construction of a drug-coordinated cell therapy system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:临床上,激素替代疗法(HRT)是原发性卵巢功能不全(POI)的主要治疗手段。然而,HRT可能会增加患乳腺癌和心血管疾病的风险。人脐带间充质干细胞(hUC-MSC)来源的外泌体通过抑制炎症反应逐渐被应用于多种疾病的治疗,免疫调节,和组织修复功能。然而,hUC-MSC外泌体在POI中的应用和研究仍然有限。
    方法:这里,我们首先构建了四种大鼠动物模型:POI-C模型(“环磷酰胺诱导的”腹腔注射POI模型),POI-B模型(“白消安诱导”POI模型),POI-U模型(超声引导下的“环磷酰胺诱导”POI模型),和MS模型(“母体分离模型”)。第二,我们比较了体重,卵巢指数,status,大鼠恶情量表,并发症,不同POI大鼠模型的死亡率。最后,经腹超声引导注射hUC-MSC外泌体,并评价其对POI动物模型的治疗效果,包括激素水平的变化,发情周期,卵巢细胞凋亡水平,和生育能力。此外,我们进行RNA-seq以探索hUC-MSC外泌体功能的可能机制。
    结果:与POI-C相比,POI-B,和MS动物模型,POI-U模型显示重量波动较小,较低的卵巢指数,并发症少,较低的死亡率,和更高的模型成功率。第二,我们成功鉴定了hUC-MSCs及其外泌体,并进行超声引导下卵巢内注射hUC-MSCs外泌体。最后,我们证实超声引导的外泌体注射(称为POI-e)有效地改善了卵巢激素水平,发情周期,卵巢功能,和生育能力。机械上,hUC-MSCs可能通过调节卵巢免疫和代谢功能发挥治疗作用。
    结论:在我们的研究中,我们创新性地构建了超声引导下卵巢药物注射方法,构建POI-U动物模型和hUC-MSC外泌体注射.并且我们证实了hUC-MSC外泌体对POI-U动物模型的治疗效果。我们的研究将为POI的新型动物模型提供更好的选择,并为POI患者的hUC-MSCs外泌体治疗提供一定的指导。
    BACKGROUND: Clinically, hormone replacement therapy (HRT) is the main treatment for primary ovarian insufficiency (POI). However, HRT may increase the risk of both breast cancer and cardiovascular disease. Exosomes derived from human umbilical cord mesenchymal stem cell (hUC-MSC) have been gradually applied to the therapy of a variety of diseases through inflammation inhibition, immune regulation, and tissue repair functions. However, the application and study of hUC-MSC exosomes in POI remain limited.
    METHODS: Here, we first constructed four rat animal models: the POI-C model (the \"cyclophosphamide-induced\" POI model via intraperitoneal injection), the POI-B model (the \"busulfan-induced\" POI model), the POI-U model (the \"cyclophosphamide-induced\" POI model under ultrasonic guidance), and MS model (the \"maternal separation model\"). Second, we compared the body weight, ovarian index, status, Rat Grimace Scale, complications, and mortality rate of different POI rat models. Finally, a transabdominal ultrasound-guided injection of hUC-MSC exosomes was performed, and its therapeuticy effects on the POI animal models were evaluated, including changes in hormone levels, oestrous cycles, ovarian apoptosis levels, and fertility. In addition, we performed RNA-seq to explore the possible mechanism of hUC-MSC exosomes function.
    RESULTS: Compared with the POI-C, POI-B, and MS animal models, the POI-U model showed less fluctuation in weight, a lower ovarian index, fewer complications, a lower mortality rate, and a higher model success rate. Second, we successfully identified hUC-MSCs and their exosomes, and performed ultrasound-guided intraovarian hUC-MSCs exosomes injection. Finally, we confirmed that the ultrasound-guided exosome injection (termed POI-e) effectively improved ovarian hormone levels, the oestrous cycle, ovarian function, and fertility. Mechanically, hUC-MSCs may play a therapeutic role by regulating ovarian immune and metabolic functions.
    CONCLUSIONS: In our study, we innovatively constructed an ultrasound-guided ovarian drug injection method to construct POI-U animal models and hUC-MSC exosomes injection. And we confirmed the therapeutic efficacy of hUC-MSC exosomes on the POI-U animal models. Our study will offer a better choice for new animal models of POI in the future and provides certain guidance for the hUC-MSCs exosome therapy in POI patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    反义寡核苷酸(ASO)是靶向治疗癌症的新型治疗平台。以前,我们已经证明miR-146b-5p在结直肠癌进展中起重要作用.然而,将ASO递送到其靶向RNA的安全有效策略仍然是翻译进展的主要障碍。人脐带间充质细胞(hUC-MSC)来源的外来体用作载体以递送抗miR-146b-5pASO(PMO-146b)。PMO-146b通过共价缀合到识别外泌体表面标记的锚定肽CP05(P)上组装到外泌体(e)的表面上。CD63,形成名为ePPMO-146b的复合物。在ePPMO-146b治疗后,细胞增殖,吸收能力,并进行了迁移测定,并在体外评估上皮-间质转化的进展。使用小鼠异种移植模型来确定ePPMO-146b在体内的抗肿瘤作用和分布。ePPMO-146b被SW620细胞吸收并有效抑制细胞增殖和迁移。该缀合物还通过系统给药在结肠癌的异种移植小鼠模型中发挥抗肿瘤功效。其中PPMO-146b在肿瘤组织中富集。我们的研究强调了用PPMO-146b锚定的hUC-MSC衍生的外泌体作为PMO支持的ASO递送的新型安全有效方法的潜力。
    Antisense oligonucleotide (ASO) is a novel therapeutic platform for targeted cancer therapy. Previously, we have demonstrated that miR-146b-5p plays an important role in colorectal cancer progression. However, a safe and effective strategy for delivery of an ASO to its targeted RNA remains as a major hurdle in translational advances. Human umbilical cord mesenchymal cell (hUC-MSC)-derived exosomes were used as vehicles to deliver an anti-miR-146b-5p ASO (PMO-146b). PMO-146b was assembled onto the surface of exosomes (e) through covalent conjugation to an anchor peptide CP05 (P) that recognized an exosomal surface marker, CD63, forming a complex named ePPMO-146b. After ePPMO-146b treatment, cell proliferation, uptake ability, and migration assays were performed, and epithelial-mesenchymal transition progression was evaluated in vitro. A mouse xenograft model was used to determine the antitumor effect and distribution of ePPMO-146b in vivo. ePPMO-146b was taken up by SW620 cells and effectively inhibited cell proliferation and migration. The conjugate also exerted antitumor efficacy in a xenograft mouse model of colon cancer by systematic administration, where PPMO-146b was enriched in tumor tissue. Our study highlights the potential of hUC-MSC-derived exosomes anchored with PPMO-146b as a novel safe and effective approach for PMO backboned ASO delivery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:人脐带间充质干细胞(hUC-MSC)移植治疗系统性红斑狼疮(SLE)的疗效已被小规模临床试验证实。然而,这些试验集中在严重或难治性SLE,虽然很少有研究集中在轻度SLE。因此,本研究集中于hUC-MSC移植对早期或轻度MRL/lpr狼疮模型小鼠的治疗作用。
    方法:通过尾静脉注射将市售hUC-MSC移植到8周龄MRL/lpr小鼠中。流式细胞术用于分析外周血中的B细胞及其亚群。Further,血浆炎症因子,自身抗体,采用蛋白芯片技术和ELISA试剂盒检测血浆生化指标。此外,病理染色和免疫荧光检测小鼠肾损伤。
    结果:hUC-MSC移植对小鼠体重无影响,中剂量和高剂量hUC-MSC移植(MD和HD组)实际上降低了脾脏重量。hUC-MSC移植显着降低了浆母细胞(PB)的比例,IgG1-PB,IgG1+PB,IgG1+记忆B(MB)细胞,IgG1+DNMB,和IgG1+SPMB细胞。hUC-MSC移植后血浆炎症因子水平明显降低,如TNF-α,IFN-γ,IL-6和IL-13。病理染色显示,hUC-MSC移植小鼠肾小球炎症细胞浸润明显减轻,肾小球纤维化水平明显减轻。免疫荧光分析表明,hUC-MSC移植小鼠肾脏中IgG和IgM抗体的沉积明显低于对照组。
    结论:hUC-MSC移植能抑制MRL/lpr小鼠早期外周血B细胞的增殖和分化,从而减轻小鼠的血浆炎症环境,导致肾损伤缓解。该研究为SLE的治疗提供了新的、可行的策略。
    The efficacy of human umbilical cord mesenchymal stem cell (hUC-MSC) transplantation in treating systemic lupus erythematosus (SLE) has been confirmed by small-scale clinical trials. However, these trials focused on severe or refractory SLE, while few studies focused on mild SLE. Therefore, this study focused on the therapeutic effects of hUC-MSC transplantation in early-stage or mild MRL/lpr lupus model mice.
    Commercially available hUC-MSCs were transplanted into 8-week-old MRL/lpr mice by tail vein injection. Flow cytometry was used to analyze B cells and their subsets in the peripheral blood. Further, plasma inflammatory factors, autoantibodies, and plasma biochemical indices were detected using protein chip technology and ELISA kits. In addition, pathological staining and immunofluorescence were performed to detect kidney injury in mice.
    hUC-MSC transplantation did not affect the mice\'s body weight, and both middle and high dose hUC-MSC transplantation (MD and HD group) actually reduced spleen weight. hUC-MSC transplantation significantly decreased the proportion of plasmablasts (PB), IgG1- PB, IgG1+ PB, IgG1+ memory B (MB) cells, IgG1+ DN MB, and IgG1+ SP MB cells. The hUC-MSC transplantation had significantly reduced plasma levels of inflammatory factors, such as TNF-α, IFN-γ, IL-6, and IL-13. Pathological staining showed that the infiltration of glomerular inflammatory cells was significantly reduced and that the level of glomerular fibrosis was significantly alleviated in hUC-MSC-transplanted mice. Immunofluorescence assays showed that the deposition of IgG and IgM antibodies in the kidneys of hUC-MSC-transplanted mice was significantly lower than in the control.
    hUC-MSC transplantation could inhibit the proliferation and differentiation of peripheral blood B cells in the early-stage of MRL/lpr mice, thereby alleviating the plasma inflammatory environment in mice, leading to kidney injury remission. The study provides a new and feasible strategy for SLE treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    间充质干细胞/基质细胞(MSC)是梭形样异质细胞群,具有有利的双向免疫调节和造血支持作用。据报道,血管细胞粘附分子-1(VCAM-1)+MSC表现出免疫调节和促血管生成能力。这里,我们研究了VCAM-1+人脐带(hUC)-MSCs对脑梗死的神经保护作用。Sprague-Dawley大鼠进行大脑中动脉闭塞(MCAO),在MCAO手术后4小时,将VCAM-1和VCAM-1+hUC-MSC静脉注射到大鼠体内。此后,确定改良的神经系统严重度评分(mNSS),和莫里斯水迷宫测试,氯化2,3,5-三苯基四唑(TTC),苏木精和曙红(H&E),Nissl,TUNEL染色,并进行qRT-PCR。在诱导氧-葡萄糖剥夺/复氧后(OGD/R),将SH-SY5Y细胞与VCAM-1和VCAM-1+hUC-MSC共培养。CCK-8,流式细胞术,ELISA,和western印迹分析在体外进行。与VCAM-1-hUC-MSCs相比,VCAM-1+hUC-MSCs的给药显示对大鼠脑梗死的治疗效果改善,更低的mNSS评分和梗死体积证实了这一点,以及提高学习和记忆能力。此外,VCAM-1+hUC-MSCs对脑梗死大鼠神经功能缺损有改善作用,伴随着NLRP3介导的炎症反应的抑制。VCAM-1+hUC-MSC共培养改善了OGD/R处理的SH-SY5Y细胞中的活力并减少了NLRP3介导的炎症反应。此外,NLRP3在SH-SY5Y细胞中的过表达阻止了VCAM-1+hUC-MSC共培养的有益作用。总的来说,我们的研究结果证明了基于VCAM-1+hUC-MSC的细胞治疗与针对脑梗死的临床前神经保护的相关性.
    Mesenchymal stem/stromal cells (MSCs) are spindle-like heterogeneous cell populations with advantageous bidirectional immunomodulatory and hematopoietic support effects. Vascular cellular adhesion molecule-1 (VCAM-1)+ MSCs have been reported to exhibit immunoregulatory and proangiogenic capacities. Here, we studied the effects of VCAM-1+ human umbilical cord (hUC)-MSCs on neuroprotection against cerebral infarction. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO), and VCAM-1- and VCAM-1+ hUC-MSCs were intravenously injected into the rat 4 h post-MCAO surgery. Thereafter, modified neurological severity scores (mNSS) were determined, and the Morris water maze test, 2,3,5-triphenyltetrazolium chloride (TTC), hematoxylin and eosin (H&E), Nissl, TUNEL staining, and qRT-PCR were conducted. Following induction of oxygen-glucose deprivation/reoxygenation (OGD/R), SH-SY5Y cells were co-cultured with VCAM-1- and VCAM-1+ hUC-MSCs. CCK-8, flow cytometry, ELISA, and western blot analyses were performed in vitro. Compared with VCAM-1- hUC-MSCs, administration of VCAM-1+ hUC-MSCs revealed improved therapeutic efficacy against cerebral infarction in rats, as confirmed by lower mNSS scores and infarct volumes, as well as improved learning and memory capacities. In addition, VCAM-1+ hUC-MSCs exhibited improved efficacy against neurological defects in rats with cerebral infarction, accompanied by inhibition of the NLRP3-mediated inflammatory response. VCAM-1+ hUC-MSC co-culture improved the viability and diminished NLRP3-mediated inflammatory response in OGD/R-treated SH-SY5Y cells. Moreover, NLRP3 overexpression in SH-SY5Y cells prevented the beneficial effects of VCAM-1+ hUC-MSC co-culture. Overall, our findings demonstrated the relevance of VCAM-1+ hUC-MSC-based cytotherapy for preclinical neuroprotection against cerebral infarction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:阻止或逆转肝纤维化的有效疗法尚未出现,因为这些潜在药物不能特异性靶向激活的肝星状细胞(aHSC)或经常对实质细胞有毒性。人脐带间充质干细胞(Huc-MSC)来源的外泌体在纳米医学中显示出治疗肝纤维化的前景。然而,全身注射显示未修饰的外泌体主要被单核吞噬细胞系统吸收。选择性结合特定靶标的配体的发现在临床相关的诊断和治疗中起着至关重要的作用。在这里,我们旨在通过筛选噬菌体展示肽文库来鉴定aHSC的靶向肽,并用靶向肽修饰Huc-MSC衍生的外泌体。
    结果:在这项研究中,我们通过生物淘选筛选了噬菌体展示肽文库中优先结合HSC-T6细胞的肽。鉴定的肽,HSTP1在纤维化肝组织的病理切片中也表现出对aHSC的更好靶向能力。然后,HSTP1与富含外泌体的膜蛋白(Lamp2b)融合,并通过基因工程技术在外泌体表面展示。工程化外泌体(HSTP1-Exos)可以被HSC-T6细胞更有效地内化,并且在增强外泌体促进HSC-T6回复为静止表型的能力方面优于未修饰的外泌体(Blank-Exos)和Lamp2b蛋白过表达的外泌体(Lamp2b+Exos)。体内结果显示HSTP1-Exos在静脉给药后可以特异性靶向aHSC区域,如通过与典型的aHSCs标记α-SMA的共免疫荧光所证明的,增强对肝纤维化的治疗作用。
    结论:这些结果表明HSTP1是一种可靠的靶向肽,可以特异性结合aHSC,并且HSTP1修饰的外泌体实现了复杂肝组织中aHSC的精确治疗。我们为临床肝纤维化治疗提供了一种新的策略。
    BACKGROUND: Effective therapeutics to stop or reverse liver fibrosis have not emerged, because these potential agents cannot specifically target activated hepatic stellate cells (aHSCs) or are frequently toxic to parenchymal cells. Human umbilical cord mesenchymal stem cell (Huc-MSC)-derived exosomes show promise in nanomedicine for the treatment of liver fibrosis. However, systemic injection showed that unmodified exosomes were mainly taken up by the mononuclear phagocyte system. The discovery of ligands that selectively bind to a specific target plays a crucial role in clinically relevant diagnostics and therapeutics. Herein, we aimed to identify the targeting peptide of aHSCs by screening a phage-displayed peptide library, and modify Huc-MSC-derived exosomes with the targeting peptide.
    RESULTS: In this study, we screened a phage-displayed peptide library by biopanning for peptides preferentially bound to HSC-T6 cells. The identified peptide, HSTP1, also exhibited better targeting ability to aHSCs in pathological sections of fibrotic liver tissues. Then, HSTP1 was fused with exosomal enriched membrane protein (Lamp2b) and was displayed on the surface of exosomes through genetic engineering technology. The engineered exosomes (HSTP1-Exos) could be more efficiently internalized by HSC-T6 cells and outperformed both unmodified exosomes (Blank-Exos) and Lamp2b protein overexpressed exosomes (Lamp2b + Exos) in enhancing the ability of exosomes to promote HSC-T6 reversion to a quiescent phenotype. In vivo results showed HSTP1-Exos could specifically target to the aHSC region after intravenous administration, as demonstrated by coimmunofluorescence with the typical aHSCs marker α-SMA, and enhance the therapeutic effect on liver fibrosis.
    CONCLUSIONS: These results suggest that HSTP1 is a reliable targeting peptide that can specifically bind to aHSCs and that HSTP1-modified exosomes realize the precise treatment for aHSCs in complex liver tissue. We provide a novel strategy for clinical liver fibrosis therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    未经证实:与其他类型的烧伤伤口相比,冷烧伤伤口的病理生理谱有所不同。这些伤口对身体有长期的破坏性影响,包括肥厚性疤痕,挛缩,和坏死。间充质干细胞(MSC)被认为是烧伤伤口完全再生的有希望的候选者。然而,移植的MSCs面临着在苛刻的组织条件下存活的挑战。用生物活性化合物对MSC进行预处理可以增强其存活和用于临床应用的再生潜力。马尾草的生物活性化合物因其抗炎作用而在治疗不同类型的皮肤伤口中具有潜在的作用,抗病毒,抗细胞毒性,和抗氧化性能。本研究旨在评估人脐带来源的MSCs(hUC-MSCs)在用M.azedarach生物活性化合物(槲皮素和芦丁)预处理后对冷诱导烧伤伤口的协同作用。
    UNASSIGNED:人脐带MSC(hUC-MSC)基于其特异性细胞表面标志物进行表征,并用20μM槲皮素或芦丁处理。进行体外划痕测定以测量细胞迀移和伤口闭合。通过将大鼠背部皮肤直接暴露于液氮来建立体内冷烧伤伤口模型。在烧伤伤口诱导的第二天皮下移植hUC-MSC,并在对应于伤口愈合阶段的不同时间点(第3、7和14天)检查伤口。在不同组中评估预调理hUC-MSCs的再生潜力;对照组(仅用hUC-MSCs处理),和处理组(槲皮素或芦丁处理的hUC-MSC)。通过组织学评估愈合潜力和伤口闭合,基因表达,和治疗前后伤口组织的免疫组织化学分析。
    UNASSIGNED:与对照组相比,在处理组中划痕测定显示出增强的细胞向伤口闭合迁移。伤口的宏观检查显示,对照组在第14天形成结痂,而在治疗组中,结痂脱落,伤口组织明显重塑。治疗组之间的比较显示,与用芦丁治疗的MSC相比,用槲皮素治疗的烧伤伤口显著增加愈合潜力。组织学发现显示槲皮素组皮肤层和毛囊的再生增强,而两个治疗组的新生血管形成均增加。伤口愈合介质的基因谱说明IL-5,IL-4,GPX-7,TXNRD-2,PRDX,VEGF,和FGF以及炎症细胞因子IL-1β和IL-6的下调。
    未经批准:总而言之,hUC-MSCs和M.azedarach生物活性化合物的协同作用通过减少炎症促进伤口愈合,减轻氧化应激和增强新生血管。研究结果将有助于为冷烧伤伤口设计更有效的治疗方案。
    UNASSIGNED: Cold burn wounds differ in their pathophysiological spectrum as compared to other types of burn wounds. These wounds have prolonged devastating effects on the body including hypertrophic scars, contracture, and necrosis. Mesenchymal stem cells (MSCs) are considered promising candidates for the complete regeneration of burn wounds. However, transplanted MSCs face the challenge to survive under the harsh tissue conditions. Preconditioning of MSCs with bioactive compounds may enhance their survival and regenerative potential for use in clinical applications. Bioactive compounds of Melia azedarach are well known for their potential role in treating different types of skin wounds due to their anti-inflammatory, anti-viral, anti-cytotoxic, and anti-oxidative properties. This study aims to evaluate the synergistic effects of human umbilical cord derived MSCs (hUC-MSCs) after preconditioning them with bioactive compounds of M. azedarach (quercetin and rutin) for cold induced burn wounds.
    UNASSIGNED: Human umbilical cord MSCs (hUC-MSCs) were characterized based on their specific cell surface markers and treated with 20 μM of quercetin or rutin. In vitro scratch assay was performed to measure cell migration and wound closure. In vivo cold burn wound model was developed via direct exposure of the dorsal rat skin to liquid nitrogen. hUC-MSCs were subcutaneously transplanted next day of burn wound induction and wound was examined at different time points corresponding to the wound healing phases (days 3, 7, and 14). The regenerative potential of preconditioned hUC-MSCs was assessed in different groups; control (treated only with hUC-MSCs), and treated groups (quercetin or rutin treated hUC-MSCs). Healing potential and wound closure were evaluated by histological, gene expression, and immunohistochemical analyses of the wound tissues before and after treatment.
    UNASSIGNED: Scratch assay exhibited enhanced cell migration towards wound closure in the treated groups as compared to the control. Macroscopic examination of the wound revealed scab formation at day 14 in control, whereas scab was detached and the wound tissue was remarkably remodeled in the treated groups. Comparison between the treated groups showed that burn wound treated with quercetin significantly increased healing potential than the rutin treated MSCs. Histological findings showed enhanced regeneration of skin layers along with hair follicles in the quercetin group, while increased neovascularization was noted in both treatment groups. Gene profile of wound healing mediators illustrated significant upregulation of IL-5, IL-4, GPX-7, TXNRD-2, PRDX, VEGF, and FGF and downregulation of inflammatory cytokines IL-1β and IL-6.
    UNASSIGNED: In conclusion, synergistic effect of hUC-MSCs and bioactive compounds of M. azedarach enhances wound healing by reducing the inflammation, mitigating oxidative stress and enhancing neovascularization. The study findings will aid in designing more effective treatment options for cold burn wounds.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在之前的研究中,我们已经证明,来自人脐带间充质干细胞(hUC-MSCs)的外泌体miR-451可以减轻烧伤诱导的急性肺损伤(ALI)。然而,外泌体miR-451在ALI中的作用机制尚不清楚.因此,本研究旨在研究hUC-MSCs来源的外泌体miR-451通过调节巨噬细胞极化对ALI的分子机制。通过透射电子显微镜(TEM)和纳米颗粒跟踪分析(NTA)分离和鉴定外泌体。qRT-PCR和westernblot检测miR-451、巨噬细胞移动抑制因子(MIF)和PI3K/AKT信号通路蛋白的表达。流式细胞术检测CD80和CD206阳性细胞。建立重度烧伤大鼠模型,HE检测炎性细胞浸润及炎性损伤。双荧光素酶报告系统用于检测miR-451对MIF的调控。用ELISA法检测细胞因子的含量。结果表明,hUC-MSCs外泌体促进巨噬细胞M1向M2极化。此外,hUC-MSCs来源的外泌体miR-451减轻ALI发育并促进巨噬细胞M1向M2极化。此外,MIF是miR-451的直接靶标。miR-451调控的MIF下调减轻了ALI的发育,促进了巨噬细胞M1向M2的极化。此外,我们发现MIF和hUC-MSCs来源的外泌体miR-451通过调节PI3K/AKT信号通路参与ALI.总之,我们表明hUC-MSCs来源的外泌体miR-451通过调节MIF-PI3K-AKT信号通路调节巨噬细胞M2极化减轻ALI,为烧伤后ALI的治疗提供了重要的科学意义和临床应用价值。
    In the previous study, we have proved that exosomal miR-451 from human umbilical cord mesenchymal stem cells (hUC-MSCs) attenuated burn-induced acute lung injury (ALI). However, the mechanism of exosomal miR-451 in ALI remains unclear. Therefore, this study aimed to study the molecular mechanism of hUC-MSCs-derived exosomal miR-451 on ALI by regulating macrophage polarization. Exosomes were isolated and identified by transmission electron microscope (TEM) and nanoparticle tracking analysis (NTA). The expression of miR-451, macrophage migration inhibitory factor (MIF) and PI3K/AKT signaling pathway proteins were detected by qRT-PCR and western blot. Flow cytometry was used to detect the CD80 and CD206 positive cells. Severe burn rat model was established and HE was used to detect the inflammatory cell infiltration and inflammatory injury. Dual luciferase reporter system was used to detect the regulation of miR-451 to MIF. The contents of cytokines were detected by ELISA. The results showed that hUC-MSCs exosomes promoted macrophage M1 to M2 polarization. Furthermore, hUC-MSCs-derived exosomal miR-451 alleviated ALI development and promoted macrophage M1 to M2 polarization. Moreover, MIF was a direct target of miR-451. Downregulation of MIF regulated by miR-451 alleviated ALI development promoted macrophage M1 to M2 polarization. In addition, we found that MIF and hUC-MSCs-derived exosomal miR-451 participated in ALI by regulating PI3K/AKT signaling pathway. In conclusion, we indicated that hUC-MSCs-derived exosomal miR-451 alleviated ALI by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway, which provided great scientific significance and clinical application value for the treatment of burn-induced ALI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号