Lipid nanoparticle

脂质纳米粒
  • 文章类型: Journal Article
    重度抑郁症(MDD)是一种普遍存在的精神障碍,显着影响社会和心理功能,但是目前没有有效的药物。环状RNA(circularRNAs,circRNAs)已被报道参与MDD的发病机理,被认为是有希望的治疗靶标。然而,针对MDD的circRNA的非基于病毒的递送策略没有被彻底研究.这里,发现circATF7IP在血浆样本中显著上调,并与MDD患者的24-汉密尔顿抑郁量表(HAMD-24)评分呈正相关.协同胺脂质纳米颗粒(SALNP)旨在通过鼻内给药将靶向cirATF7IP(si-circatF7IP)的siRNA递送到海马脑区。SALNP-si-circATF7IP的鼻内递送通过减少CD11bCD45dim小胶质细胞数量和促炎细胞因子的产生(TNF-α和IL-6),成功缓解了LPS诱导的小鼠抑郁模型中的抑郁样行为。这些结果表明cirATF7IP水平与MDD发病机制正相关,和SALNP通过鼻内给药递送si-circATF7IP是改善LPS诱导的抑郁样行为的有效策略。
    Major depressive disorder (MDD) is a prevalent mental disorder that significantly impacts social and psychological function, but no effective medication is currently available. Circular RNAs (circRNAs) have been reported to participate in the pathogenesis of MDD which are envisioned as promising therapeutic targets. However, nonviral-based delivery strategies targeting circRNA against MDD are not thoroughly investigated. Here, it is identified that circATF7IP is significantly upregulated in plasma samples and positively correlated with 24-Hamilton Depression Scale (HAMD-24) scores of MDD patients. Synergistic amine lipid nanoparticles (SALNPs) are designed to deliver siRNA targeting circATF7IP (si-circATF7IP) into the hippocampus brain region by intranasal administration. Intranasal delivery of SALNP-si-circATF7IP successfully alleviated the depressive-like behaviors in the LPS-induced mouse depression model via decreasing CD11b+CD45dim microglia population and pro-inflammatory cytokine productions (TNF-α and IL-6). These results indicate that the level of circATF7IP positively correlates with MDD pathogenesis, and SALNP delivery of si-circATF7IP via intranasal administration is an effective strategy to ameliorate LPS-induced depressive-like behaviors.
    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
    吲哚胺-2,3-双加氧酶(IDO)1降解色氨酸,通过饮食摄入获得,进入犬尿氨酸途径的免疫调节代谢产物。IDO1的缺乏或阻断导致啮齿动物模型中自身免疫严重程度的增强和人类对发展自身免疫的易感性增加。尽管如此,利用IDO1治疗自身免疫的治疗方式仍然有限.这里,我们使用在脂质纳米颗粒(LNP)中配制的信使(m)RNA递送含有Src肉豆蔻酰化位点的人IDO1变体,以将蛋白质锚定到质膜的内表面。这种膜锚定的IDO1增加了蛋白质产量,导致代谢物变化增加,并最终改善三种T细胞介导的自身免疫模型中的疾病:实验性自身免疫性脑脊髓炎(EAE),大鼠胶原诱导性关节炎(CIA),和急性移植物抗宿主病(aGVHD)。IDO1的功效与肝表达和全身色氨酸耗竭相关。因此,在几种特征明确的自身免疫模型中,通过mRNA传递膜锚定的IDO1抑制了免疫反应。
    Indoleamine-2,3-dioxygenase (IDO)1 degrades tryptophan, obtained through dietary intake, into immunoregulatory metabolites of the kynurenine pathway. Deficiency or blockade of IDO1 results in the enhancement of autoimmune severity in rodent models and increased susceptibility to developing autoimmunity in humans. Despite this, therapeutic modalities that leverage IDO1 for the treatment of autoimmunity remain limited. Here, we use messenger (m)RNA formulated in lipid nanoparticles (LNPs) to deliver a human IDO1 variant containing the myristoylation site of Src to anchor the protein to the inner face of the plasma membrane. This membrane-anchored IDO1 has increased protein production, leading to increased metabolite changes, and ultimately ameliorates disease in three models of T cell-mediated autoimmunity: experimental autoimmune encephalomyelitis (EAE), rat collagen-induced arthritis (CIA), and acute graft-versus-host disease (aGVHD). The efficacy of IDO1 is correlated with hepatic expression and systemic tryptophan depletion. Thus, the delivery of membrane-anchored IDO1 by mRNA suppresses the immune response in several well-characterized models of autoimmunity.
    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
    多功能疗法已成为癌症治疗的创新策略。在这篇研究文章中,我们提出了一种纳米结构的脂质载体(NLC)设计用于皮肤黑色素瘤的局部治疗,同时递送5-FU和Bcl-2siRNA。表征的纳米颗粒表现出259±9nm的直径和0.2的多分散指数,表明均匀的尺寸分布。NLC主要位于表皮,有效地减少5-FU在整个皮肤层的全身释放。离体皮肤模型揭示了保护性脂质膜的形成,减少角质层的脱皮过程,这可能与增加渗透的作用有关。体外实验表明,与非癌细胞相比,A375黑色素瘤细胞对治疗表现出更高的敏感性。反映了他们代谢率的预期差异。A375细胞对NLC的摄取在4小时内达到约90%。使用ELISA彻底评估Bcl-2敲低的功效,蛋白质印迹,和qRT-PCR分析,揭示了含有5-FU和Bcl-2siRNA(在低浓度-100μM)的NLC制剂的显著敲除和协同作用。值得注意的是,Bcl-2mRNA的沉默也影响了Bcl-2蛋白家族的其他成员,包括Mcl-1,Bcl-xl,巴克斯,还有BAK.观察到的这些蛋白质的调节强烈表明凋亡途径的激活,表明成功抑制了黑色素瘤的生长并防止了其在体外的传播。
    Multifunctional therapies have emerged as innovative strategies in cancer treatment. In this research article, we proposed a nanostructured lipid carrier (NLC) designed for the topical treatment of cutaneous melanoma, which simultaneously delivers 5-FU and Bcl-2 siRNA. The characterized nanoparticles exhibited a diameter of 259 ± 9 nm and a polydispersion index of 0.2, indicating a uniform size distribution. The NLCs were primarily localized in the epidermis, effectively minimizing the systemic release of 5-FU across skin layers. The ex vivo skin model revealed the formation of a protective lipid film, decreasing the desquamation process of the stratum corneum which can be associated to an effect of increasing permeation. In vitro assays demonstrated that A375 melanoma cells exhibited a higher sensitivity to the treatment compared to non-cancerous cells, reflecting the expected difference in their metabolic rates. The uptake of NLC by A375 cells reached approximately 90% within 4 h. The efficacy of Bcl-2 knockdown was thoroughly assessed using ELISA, Western blot, and qRT-PCR analyses, revealing a significant knockdown and synergistic action of the NLC formulation containing 5-FU and Bcl-2 siRNA (at low concentration --100 pM). Notably, the silencing of Bcl-2 mRNA also impacted other members of the Bcl-2 protein family, including Mcl-1, Bcl-xl, BAX, and BAK. The observed modulation of these proteins strongly indicated the activation of the apoptosis pathway, suggesting a successful inhibition of melanoma growth and prevention of its in vitro spread.
    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
    胰腺癌的肿瘤微环境(TME)具有高度免疫抑制性,其特征是大量与癌症相关的成纤维细胞,骨髓来源的抑制细胞,和调节性T细胞。干扰素基因刺激因子(STING)是一种内质网受体,在免疫中起着至关重要的作用。STING激动剂已经证明了煽动TME的能力,减轻肿瘤负担,并在小鼠模型中赋予抗肿瘤活性。2'3'环磷酸鸟苷一磷酸腺苷(2'3'-cGAMP)是STING的高亲和力内源性配体。然而,将cGAMP递送至细胞质内的抗原呈递细胞和肿瘤细胞仍然具有挑战性,这归因于膜不渗透性和差的稳定性。
    在这项研究中,我们将2'3'-cGAMP封装在设计用于有效细胞递送的脂质纳米颗粒(cGAMP-LNP)中。我们使用一系列旨在评估其细胞摄取的体外研究来评估纳米颗粒的特性,胞质释放,和最小的细胞毒性。此外,我们研究了纳米颗粒在同系小鼠胰腺癌模型中的抗肿瘤作用。
    脂质平台显着增加了2'3'-cGAMP的细胞摄取。cGAMP-LNP在胰腺癌的同基因小鼠模型中显示出有希望的抗肿瘤活性。
    LNP平台有望在癌症治疗中提供外源性2'3'-cGAMP或其衍生物。
    UNASSIGNED: The tumor microenvironment (TME) of pancreatic cancer is highly immunosuppressive and characterized by a large number of cancer-associated fibroblasts, myeloid-derived suppressor cells, and regulatory T cells. Stimulator of interferon genes (STING) is an endoplasmic reticulum receptor that plays a critical role in immunity. STING agonists have demonstrated the ability to inflame the TME, reduce tumor burden, and confer anti-tumor activity in mouse models. 2\'3\' cyclic guanosine monophosphate adenosine monophosphate (2\'3\'-cGAMP) is a high-affinity endogenous ligand of STING. However, delivering cGAMP to antigen-presenting cells and tumor cells within the cytosol remains challenging due to membrane impermeability and poor stability.
    UNASSIGNED: In this study, we encapsulated 2\'3\'-cGAMP in a lipid nanoparticle (cGAMP-LNP) designed for efficient cellular delivery. We assessed the properties of the nanoparticles using a series of in-vitro studies designed to evaluate their cellular uptake, cytosolic release, and minimal cytotoxicity. Furthermore, we examined the nanoparticle\'s anti-tumor effect in a syngeneic mouse model of pancreatic cancer.
    UNASSIGNED: The lipid platform significantly increased the cellular uptake of 2\'3\'-cGAMP. cGAMP-LNP exhibited promising antitumor activity in the syngeneic mouse model of pancreatic cancer.
    UNASSIGNED: The LNP platform shows promise for delivering exogenous 2\'3\'-cGAMP or its derivatives in cancer therapy.
    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
    上皮细胞粘附分子(EpCAM)基因编码I型跨膜糖蛋白,该糖蛋白在许多癌性上皮细胞中过度表达,并通过调节几种癌基因如c-myc和其他细胞周期蛋白的表达来促进肿瘤进展。由于这种致瘤性关联,最近几天,EpCAM基因已成为抗癌治疗的潜在靶点.在这里,尝试通过由合成的刺激敏感脂质制成的脂质纳米颗粒系统,有效地递送一体化的定期间隔短回文重复序列(CRISPR)质粒,从而敲除原癌基因EpCAM表达。质粒具有靶向EpCAM基因的向导RNA形式的必需信息。适配体修饰的系统选择性地靶向EpCAM过表达的细胞并有效地抑制遗传表达。它探索了开发的脂质纳米颗粒的pH响应特性,并监测了它们在不同来源的各种癌细胞系中的功效,并提高了EpCAM水平。该现象已在非免疫受损小鼠肿瘤模型中进一步得到体内验证。总的来说,新开发的适配体修饰的脂质纳米颗粒系统已被证明对于递送EpCAM靶向的CRISPR/Cas9质粒是有效的。
    Epithelial cell adhesion molecule (EpCAM) gene encodes a type-I trans-membrane glycoprotein that is overexpressed in many cancerous epithelial cells and promotes tumor progression by regulating the expression of several oncogenes like c-myc and other cyclins. Because of this tumorigenic association, the EpCAM gene has been a potential target for anti-cancer therapy in recent days. Herein, it is attempted to knockout the proto-oncogenic EpCAM expression by efficiently delivering an all-in-one Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) plasmid via a lipid nanoparticle system made out of synthetic stimuli-sensitive lipids. The plasmid possesses the necessary information in the form of a guide RNA targeted to the EpCAM gene. The aptamer decorated system selectively targets EpCAM overexpressed cells and efficiently inhibits the genetic expression. It has explored the pH-responsive property of the developed lipid nanoparticles and monitored their efficacy in various cancer cell lines of different origins with elevated EpCAM levels. The phenomenon has further been validated in vivo in non-immunocompromised mouse tumor models. Overall, the newly developed aptamer decorated lipid nanoparticle system has been proven to be efficacious for the delivery of EpCAM-targeted CRISPR/Cas9 plasmid.
    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
    冠状病毒(CoV)是人类和其他脊椎动物的重要病原体,引起严重的呼吸道和肠道感染,由于动物和人类之间的种间传播的可能性,这些感染已经对公共健康构成威胁。因此,安全的发展,有效的疫苗仍然是控制CoV感染的重中之重。以信使RNA(mRNA)为特征的疫苗的独特免疫学特征为冠状病毒疫苗开发提供了有利的工具。这里,我们设计了两种脂质纳米颗粒(LNP)包裹的mRNA(mRNA-LNP)疫苗:一种编码全长刺突(S)蛋白,另一种编码来自猪三角洲冠状病毒(PDCoV)的刺突胞外域(Se)。初次免疫后14天,两种mRNA疫苗在小鼠中诱导高水平的免疫球蛋白G和中和抗体,S疫苗表现出比Se疫苗更好的性能。通过诱导强大的PDCoV特异性体液和细胞免疫反应,证实了SmRNA疫苗在乳猪中的被动免疫保护作用。SmRNA疫苗也显示出比灭活疫苗更好的保护效果。我们的结果表明,新型PDCoV-SmRNA-LNP疫苗可能具有对抗PDCoV感染的潜力。
    目的:作为一种新兴的猪肠致病性冠状病毒,猪三角洲冠状病毒(PDCoV)具有跨物种传播的潜力,引起广泛关注。信使RNA(mRNA)疫苗是对抗新出现和重新出现的传染病的一个有希望的选择,COVID-19mRNA疫苗的有效性证明了这一点。这里,我们首先证明了PDCoV-SmRNA-脂质纳米颗粒(LNP)疫苗可以在小鼠中诱导有效的体液和细胞免疫应答。通过对SmRNA疫苗在哺乳仔猪中的被动免疫保护作用的评价,证实了mRNA疫苗的保护效果优于灭活疫苗。这项研究表明,PDCoV-SmRNA-LNP疫苗可能是对抗PDCoV感染的潜在新型疫苗。
    Coronaviruses (CoVs) are important pathogens for humans and other vertebrates, causing severe respiratory and intestinal infections that have become a threat to public health because of the potential for interspecies transmission between animals and humans. Therefore, the development of safe, effective vaccines remains a top priority for the control of CoV infection. The unique immunological characteristics of vaccines featuring messenger RNA (mRNA) present an advantageous tool for coronavirus vaccine development. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines: one encoding full-length spike (S) protein and the other encoding the spike ectodomain (Se) from porcine deltacoronavirus (PDCoV). Fourteen days after primary immunization, both mRNA vaccines induced high levels of immunoglobulin G and neutralizing antibodies in mice, with the S vaccine showing better performance than the Se vaccine. Passive immune protection of the S mRNA vaccine in suckling piglets was confirmed by the induction of robust PDCoV-specific humoral and cellular immune responses. The S mRNA vaccine also showed better protective effects than the inactivated vaccine. Our results suggest that the novel PDCoV-S mRNA-LNP vaccine may have the potential to combat PDCoV infection.
    OBJECTIVE: As an emerging porcine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) has the potential for cross-species transmission, attracting extensive attention. Messenger RNA (mRNA) vaccines are a promising option for combating emerging and re-emerging infectious diseases, as evidenced by the demonstrated efficacy of the COVID-19 mRNA vaccine. Here, we first demonstrated that PDCoV-S mRNA-lipid nanoparticle (LNP) vaccines could induce potent humoral and cellular immune responses in mice. An evaluation of passive immune protection of S mRNA vaccines in suckling piglets confirmed that the protective effect of mRNA vaccine was better than that of inactivated vaccine. This study suggests that the PDCoV-S mRNA-LNP vaccine may serve as a potential and novel vaccine candidate for combating PDCoV infection.
    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
    脂质纳米颗粒是临床上最先进的核酸治疗药物递送系统,以COVID-19mRNA疫苗的成功为例。然而,它们的临床应用目前仅限于肝脏疾病和疫苗,因为它们在静脉内给药时会在肝脏中积累。为了充分利用他们的潜力,了解和解决他们的肝脏嗜性是至关重要的,同时还制定策略以增强向肝脏以外组织的输送。确保这些治疗剂到达它们的靶细胞同时避免脱靶细胞对于它们的功效和安全性都是至关重要的。有三种潜在的瞄准策略-被动,活跃,和内源性-其可以单独或组合用于靶向非肝组织。在这次审查中,我们将深入研究脂质纳米颗粒工程的最新进展,以将核酸递送到肝脏之外。
    Lipid nanoparticles (LNPs) are the most clinically advanced drug delivery system for nucleic acid therapeutics, exemplified by the success of the COVID-19 mRNA vaccines. However, their clinical use is currently limited to hepatic diseases and vaccines due to their tendency to accumulate in the liver upon intravenous administration. To fully leverage their potential, it is essential to understand and address their liver tropism, while also developing strategies to enhance delivery to tissues beyond the liver. Ensuring that these therapeutics reach their target cells while avoiding off-target cells is essential for both their efficacy and safety. There are three potential targeting strategies-passive, active, and endogenous-which can be used individually or in combination to target nonhepatic tissues. In this review, we delve into the recent advancements in LNP engineering for delivering nucleic acid beyond the liver.
    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
    据报道脑啡肽与心脏功能相关。然而,它们在心脏中的调节仍未被探索。这项研究表明,在接受多柔比星(Dox)治疗的受试者中,阿片样物质生长因子(OGF)(也称为甲硫氨酸脑啡肽)的循环水平以及OGF及其受体(OGFR)的心肌表达水平显着增加。通过基因敲除或使用携带小发夹RNA的腺相关病毒血清型9沉默OGFR有效地减轻了小鼠中Dox诱导的心脏毒性(DIC)。相反,补充OGF会加剧DIC表现,可以通过施用OGFR拮抗剂纳曲酮(NTX)来消除。机械上,先前表征的OGF/OGFR/P21轴被鉴定为促进DIC相关心肌细胞凋亡.此外,观察到OGFR将STAT1与铁蛋白基因(FTH和FTL)的启动子解离,从而抑制其转录并加剧DIC相关的心肌细胞铁性凋亡。为了避免由于OGFR阻断而导致的Dox对肿瘤的治疗效果受损,基于SiO2的可修饰脂质纳米颗粒被开发用于NTX的心脏靶向递送。用组装的NTX纳米药物预处理荷瘤小鼠成功地提供了针对Dox毒性的心脏保护,而不影响肿瘤中的Dox治疗。一起来看,这项研究提供了对Dox心脏毒性的新认识,并为接受Dox治疗的肿瘤患者的心脏保护剂的开发提供了启示。
    Enkephalins are reportedly correlated with heart function. However, their regulation in the heart remains unexplored. This study revealed a substantial increase in circulating levels of opioid growth factor (OGF) (also known as methionine enkephalin) and myocardial expression levels of both OGF and its receptor (OGFR) in subjects treated with doxorubicin (Dox). Silencing OGFR through gene knockout or using adeno-associated virus serotype 9 carrying small hairpin RNA effectively alleviated Dox-induced cardiotoxicity (DIC) in mice. Conversely, OGF supplementation exacerbated DIC manifestations, which could be abolished by administration of the OGFR antagonist naltrexone (NTX). Mechanistically, the previously characterized OGF/OGFR/P21 axis was identified to facilitate DIC-related cardiomyocyte apoptosis. Additionally, OGFR was observed to dissociate STAT1 from the promoters of ferritin genes (FTH and FTL), thereby repressing their transcription and exacerbating DIC-related cardiomyocyte ferroptosis. To circumvent the compromised therapeutic effects of Dox on tumors owing to OGFR blockade, SiO2-based modifiable lipid nanoparticles were developed for heart-targeted delivery of NTX. The pretreatment of tumor-bearing mice with the assembled NTX nanodrug successfully provided cardioprotection against Dox toxicity without affecting Dox therapy in tumors. Taken together, this study provides a novel understanding of Dox cardiotoxicity and sheds light on the development of cardioprotectants for patients with tumors receiving Dox treatment.
    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
    由于MMUT酶缺乏引起的分离的甲基丙二酸血症/酸尿症(MMA)是一种非常罕见的儿科疾病,具有高发病率和死亡率,没有批准的疾病改变疗法。先前的出版物表明,用编码野生型人MMUT(MMUT)的密码子优化的mRNA进行全身治疗是治疗MMA的有希望的策略。我们开发了第二代药物产品,mRNA-3705,由编码在脂质纳米颗粒(LNP)中配制的MMUT酶的mRNA组成,其中掺入了超过先前临床候选mRNA-3704的增强。静脉注射时,两种药物产品都会在大鼠肝脏中产生功能性MMUT,并在两种MMA小鼠模型中显示出长期安全性和有效性。以相同的剂量水平给予时,mRNA-3705产生的肝MMUT蛋白表达水平比第一代药物产品mRNA-3704高2.1-3.4倍,这导致血浆甲基丙二酸的更大和更持续的减少。本文提供的数据提供了全面的临床前药理学以支持mRNA-3705的临床发展。
    Isolated methylmalonic acidemia/aciduria (MMA) due to MMUT enzyme deficiency is an ultra-rare pediatric disease with high morbidity and mortality, with no approved disease-altering therapies. Previous publications showed that systemic treatment with a codon-optimized mRNA encoding wild-type human MMUT (MMUT) is a promising strategy for treatment of MMA. We developed a second-generation drug product, mRNA-3705, comprised of an mRNA encoding the MMUT enzyme formulated in a lipid nanoparticle (LNP) with incorporation of enhancements over the previous clinical candidate mRNA-3704. Both drug products produced functional MMUT in rat livers when dosed IV, and showed long-term safety and efficacy in two mouse models of MMA. mRNA-3705 produced 2.1-3.4-fold higher levels of hepatic MMUT protein expression than the first-generation drug product mRNA-3704 when given at an identical dose level, which resulted in greater and more sustained reductions in plasma methylmalonic acid. The data presented herein provide comprehensive preclinical pharmacology to support the clinical development of mRNA-3705.
    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
    间充质干细胞(MSC)移植对各种疾病的功效已有报道。我们先前通过使用微流体装置大规模封装辅酶Q10(CoQ10)开发了靶向线粒体的药物递送系统(MITO-Porter)。当前的研究旨在确认MITO-Porter封装的CoQ10治疗是否增强了MSCs的线粒体功能,具有改善MSC移植治疗的潜力。我们使用了高纯度的人骨髓来源的MSCs,描述为快速扩增的克隆(RECs),并尝试使用微流体装置系统控制和增加封装在MITO-Porter中的CoQ10的量。我们用CoQ10封装的MITO-Porter处理了这些REC,并评估了它的细胞摄取,与线粒体共定位,线粒体呼吸能力的变化,和细胞毒性。用先前的CoQ10包裹的MITO-Porter治疗后,线粒体呼吸能力没有显着变化;但是,富含辅酶Q10的MITO-Porter治疗可显著增加RECs的线粒体呼吸容量.利用微流体装置可以控制封装在MITO-Porter中的CoQ10的量,用富含CoQ10的MITO-Porter治疗成功地激活了MSCs中的线粒体功能。因此,MITO-Porter系统提供了改善MSC细胞移植治疗的有前景的工具。
    The efficacy of mesenchymal stem cell (MSC) transplantation has been reported for various diseases. We previously developed a drug delivery system targeting mitochondria (MITO-Porter) by using a microfluidic device to encapsulate Coenzyme Q10 (CoQ10) on a large scale. The current study aimed to confirm if treatment with CoQ10 encapsulated by MITO-Porter enhanced mitochondrial functions in MSCs, with the potential to improve MSC transplantation therapy. We used highly purified human bone marrow-derived MSCs, described as rapidly expanding clones (RECs), and attempted to control and increase the amount of CoQ10 encapsulated in the MITO-Porter using microfluidic device system. We treated these RECs with CoQ10 encapsulated MITO-Porter, and evaluated its cellular uptake, co-localization with mitochondria, changes in mitochondrial respiratory capacity, and cellular toxicity. There was no significant change in mitochondrial respiratory capacity following treatment with the previous CoQ10 encapsulated MITO-Porter; however, mitochondrial respiratory capacity in RECs was significantly increased by treatment with CoQ10-rich MITO-Porter. Utilization of a microfluidic device enabled the amount of CoQ10 encapsulated in MITO-Porter to be controlled, and treatment with CoQ10-rich MITO-Porter successfully activated mitochondrial functions in MSCs. The MITO-Porter system thus provides a promising tool to improve MSC cell transplantation therapy.
    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