targeting

Targeting
  • 文章类型: Journal Article
    锁定核酸(LNA)是反义寡核苷酸(ASO)的亚型,其特征在于糖部分内的桥。LNA归功于这种化学修饰,顾名思义,把它锁在一个构象中。这种观点包括两个组成部分:一侧对ASO的总体概述,另一侧则关注脂质纳米颗粒(LNP)的递送问题。在整个过程中,对正在进行的涉及ASO的临床试验进行了筛选,以及使用LNA的多功能性和挑战。最后,我们强调了LNP作为成功交付LNA的载体的潜力。
    Locked nucleic acids (LNAs) are a subtype of antisense oligonucleotides (ASOs) that are characterized by a bridge within the sugar moiety. LNAs owe their robustness to this chemical modification, which as the name suggests, locks it in one conformation. This perspective includes two components: a general overview on ASOs from one side and on delivery issues focusing on lipid nanoparticles (LNPs) on the other side. Throughout, a screening of the ongoing clinical trials involving ASOs is given, as well as a take on the versatility and challenges of using LNAs. Finally, we highlight the potential of LNPs as carriers for the successful delivery of LNAs.
    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
    尽管小干扰RNA(siRNA)是许多疾病相关基因的特异性沉默,它们的临床应用仍然需要安全有效的方式递送到靶细胞中。高效脂质纳米颗粒(LNPs)被开发用于siRNA递送,展示了新型pH响应性脂氨基异肽(XP)载体的优势。这些序列定义的XPs通过可阳离子化的极性琥珀酰四亚乙基五胺(Stp)单元和非极性脂氨基脂肪酸(LAF)之间的支链赖氨酸键以各种比例组装成束或U形拓扑结构。使用LAF4-Stp1XPs作为可电离化合物的siRNA-LNPs的制剂导致强大的细胞摄取,高度内体逃逸,和在极低(150皮克)siRNA剂量下成功的体外基因沉默活性。意义重大的是在静脉注射入小鼠后,具有LAF4-Stp1XP束的siRNA-LNP在体内的功能内皮嗜性,与基于DLin-MC3-DMA的LNP相比,肝窦内皮细胞(LSEC)衍生因子VIII(FVIII)的高敲低和肝细胞衍生FVII的中度沉默证明。在用配体c(RGDfK)点击修饰siRNA-LNP后优化脂质组成有效沉默肿瘤内皮细胞(TECs)中的血管内皮生长因子受体-2(VEGFR-2)。这些发现揭示了可电离的XPs在LNP体内细胞型功能靶向中的作用,为未来的治疗应用奠定基础。
    Although small-interfering RNAs (siRNAs) are specific silencers for numerous disease-related genes, their clinical applications still require safe and effective means of delivery into target cells. Highly efficient lipid nanoparticles (LNPs) are developed for siRNA delivery, showcasing the advantages of novel pH-responsive lipoamino xenopeptide (XP) carriers. These sequence-defined XPs are assembled by branched lysine linkages between cationizable polar succinoyl tetraethylene pentamine (Stp) units and apolar lipoamino fatty acids (LAFs) at various ratios into bundle or U-shape topologies. Formulation of siRNA-LNPs using LAF4-Stp1 XPs as ionizable compounds led to robust cellular uptake, high endosomal escape, and successful in vitro gene silencing activity at an extremely low (150 picogram) siRNA dose. Of significance is the functional in vivo endothelium tropism of siRNA-LNPs with bundle LAF4-Stp1 XP after intravenous injection into mice, demonstrated by superior knockdown of liver sinusoidal endothelial cell (LSEC)-derived factor VIII (FVIII) and moderate silencing of hepatocyte-derived FVII compared to DLin-MC3-DMA-based LNPs. Optimizing lipid composition following click-modification of siRNA-LNPs with ligand c(RGDfK) efficiently silenced vascular endothelial growth factor receptor-2 (VEGFR-2) in tumor endothelial cells (TECs). The findings shed light on the role of ionizable XPs in the LNP in vivo cell-type functional targeting, laying the groundwork for future therapeutic applications.
    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
    噬菌体(噬菌体)代表了一种独特的病毒类别,具有选择性感染宿主细菌的显着能力,特征在于它们从蛋白质和核酸组装。利用其特殊的生物学特性和可修改的特性,噬菌体以创新的方式出现,安全,和有效的交付载体。与传统纳米载体在药物和基因递送领域相关的潜在缺点包括缺乏细胞特异性靶向,细胞毒性,并降低体内转染效率。相比之下,工程噬菌体,当被用作货物交付载体时,承诺克服这些限制,并实现增强的交付效力。这篇综述全面概述了目前噬菌体工程的策略,描述了在药物和基因递送中用作纳米载体的噬菌体的主要类型,并探讨了基于噬菌体的递送系统在疾病治疗中的应用。此外,提供了深刻的分析,批判性地研究纳米技术领域内基于噬菌体的递送系统所面临的挑战。本文的主要目的是提供理论参考,有助于有效的基于噬菌体的递送系统的合理设计和开发。
    Bacteriophages (phages) represent a unique category of viruses with a remarkable ability to selectively infect host bacteria, characterized by their assembly from proteins and nucleic acids. Leveraging their exceptional biological properties and modifiable characteristics, phages emerge as innovative, safe, and efficient delivery vectors. The potential drawbacks associated with conventional nanocarriers in the realms of drug and gene delivery include a lack of cell-specific targeting, cytotoxicity, and diminished in vivo transfection efficiency. In contrast, engineered phages, when employed as cargo delivery vectors, hold the promise to surmount these limitations and attain enhanced delivery efficacy. This review comprehensively outlines current strategies for the engineering of phages, delineates the principal types of phages utilized as nanocarriers in drug and gene delivery, and explores the application of phage-based delivery systems in disease therapy. Additionally, an incisive analysis is provided, critically examining the challenges confronted by phage-based delivery systems within the domain of nanotechnology. The primary objective of this article is to furnish a theoretical reference that contributes to the reasoned design and development of potent phage-based delivery systems.
    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
    针对婴儿血管瘤(IHs)的靶向治疗已被广泛研究,因为它们可以浓缩药物,增加治疗效果和减少药物剂量。同时,它们可以延长药物释放时间,增强药物稳定性,减少给药频率,提高患者的依从性。此外,由生物相容性材料制成的载体降低了药物的免疫原性,尽量减少不良反应。然而,目前的靶向制剂仍面临诸多挑战,如载体材料的非绝对安全性;需要进一步增加载药能力;动物血管瘤模型在充分复制人婴儿血管瘤生物学特性方面的局限性;建立发病率高的深层血管瘤模型;以及开发更特异性的靶标或标志物。在这次审查中,我们简要概述了IHs的特点,并总结了过去十年的进展,优势,和IHs靶向给药系统的靶向策略,并讨论了它们在IHs治疗中的应用。此外,目的是为该领域的进一步研究和应用提供参考。
    Targeted therapy for infantile hemangiomas (IHs) has been extensively studied as they can concentrate drugs, increase therapeutic efficacy and reduce drug dosage. Meanwhile, they can extend drug release times, enhance drug stability, decrease dosing frequency, and improve patient compliance. Moreover, carriers made from biocompatible materials reduced drug immunogenicity, minimizing adverse reactions. However, current targeted formulations still face numerous challenges such as the non-absolute safety of carrier materials; the need to further increase drug loading capacity; the limitation of animal hemangioma models in fully replicating the biological properties of human infantile hemangiomas; the establishment of models for deep-seated hemangiomas with high incidence rates; and the development of more specific targets or markers. In this review, we provided a brief overview of the characteristics of IHs and summarized the past decade\'s advances, advantages, and targeting strategies of targeted drug delivery systems for IHs and discussed their applications in the treatment of IHs. Furthermore, the goal is to provide a reference for further research and application in this field.
    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
    树突状细胞(DC)由于其有效的抗原呈递能力而呈现用于递送免疫原性货物的理想靶标。这种靶向方法通过提高DC的抗原识别和捕获效率而在疫苗开发中具有希望。为了鉴定与兔DC结合的高亲和力靶向肽,分离并培养兔单核细胞来源的DC(raMoDC),和一种新的肽,HS(HSLRHDYGYPGH),使用噬菌体展示的肽文库鉴定。在HS旁边,另外两种DC靶向肽,KC1和MY,先前在我们的实验室中验证过,构建重组罗伊乳杆菌融合表达兔出血症病毒(RHDV)衣壳蛋白VP60。这些重组乳杆菌菌株被命名为HS-VP60/L。reuteri,KC1-VP60/Lreuteri,和MY-VP60/Lreuteri.在体内和体外评估了这些重组乳杆菌结合兔DC的能力。结果表明,DC靶向肽KC1显著提高了raMoDC对重组乳酸菌的捕获效率,促进DC成熟,和细胞因子分泌增加。此外,口服KC1-VP60/L罗伊特能有效诱导兔SIgA和IgG的产生,攻击后兔子存活时间延长,并减少器官中的RHDV拷贝。总之,DC靶向肽KC1表现出与raMoDC的强结合,表达KC1-VP60蛋白抗原的重组乳酸菌可有效诱导兔全身和粘膜免疫反应,赋予对RHDV的保护功效。这项研究为新型RHDV疫苗的开发提供了有价值的见解。
    Dendritic cells (DCs) present an ideal target for delivering immunogenic cargo due to their potent antigen-presenting capabilities. This targeting approach holds promise in vaccine development by enhancing the efficiency of antigen recognition and capture by DCs. To identify a high-affinity targeting peptide binding to rabbit DCs, rabbit monocyte-derived DCs (raMoDCs) were isolated and cultured, and a novel peptide, HS (HSLRHDYGYPGH), was identified using a phage-displayed peptide library. Alongside HS, two other DC-targeting peptides, KC1 and MY, previously validated in our laboratory, were employed to construct recombinant Lactgobacillus reuteri fusion-expressed rabbit hemorrhagic disease virus (RHDV) capsid protein VP60. These recombinant Lactobacillus strains were named HS-VP60/L. reuteri, KC1-VP60/L. reuteri, and MY-VP60/L. reuteri. The ability of these recombinant Lactobacillus to bind rabbit DCs was evaluated both in vivo and in vitro. Results demonstrated that the DC-targeting peptide KC1 significantly enhanced the capture efficiency of recombinant Lactobacillus by raMoDCs, promoted DC maturation, and increased cytokine secretion. Furthermore, oral administration of KC1-VP60/L. reuteri effectively induced SIgA and IgG production in rabbits, prolonged rabbit survival post-challenge, and reduced RHDV copies in organs. In summary, the DC-targeting peptide KC1 exhibited robust binding to raMoDCs, and recombinant Lactobacillus expressing KC1-VP60 protein antigens efficiently induced systemic and mucosal immune responses in rabbits, conferring protective efficacy against RHDV. This study offers valuable insights for the development of novel RHDV vaccines.
    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
    弓形虫具有重要的治疗潜力;然而,其非特异性侵袭性导致脱靶效应。这项研究的目的是评估弓形虫特异性是否可以通过表面展示针对树突状细胞的scFv来提高,DEC205和免疫检查点PD-L1。抗DEC205scFv直接经由糖基磷脂酰肌醇(GPI)或通过与SAG1蛋白融合而锚定至弓形虫表面。两个构建体都成功表达,但是结合结果表明,抗DEC-SAG1scFv对重组DEC蛋白和表达DEC205的MutuDC细胞具有更可靠的功能。开发了两种在HA标签的定位上不同的抗PD-L1scFv构建体。两种结构都得到了充分表达,但是HA标签的定位决定了与PD-L1蛋白结合的功能。显示抗PD-L1scFv的弓形虫与表达/显示不同水平的PD-L1的肿瘤细胞的共孵育显示强结合,这取决于可用生物标志物的水平。中和测定证实,结合是由于抗PD-L1scFv与其配体之间的特异性相互作用。混合细胞试验表明,表达抗PD-L1scFv的弓形虫主要靶向PD-L1阳性细胞,具有可忽略的脱靶结合。与亲本菌株相比,重组RH-PD-L1-C菌株对PD-L1肿瘤细胞系的杀伤能力增加。此外,靶肿瘤细胞和效应CD8+T细胞共培养试验表明,我们的模型可以抑制PD1/PD-L1相互作用并增强T细胞免疫应答.这些发现强调了抗体片段的表面展示作为靶向复制性弓形虫菌株同时最小化非特异性结合的有希望的策略。
    Toxoplasma gondii holds significant therapeutic potential; however, its nonspecific invasiveness results in off-target effects. The purpose of this study is to evaluate whether T. gondii specificity can be improved by surface display of scFv directed against dendritic cells\' endocytic receptor, DEC205, and immune checkpoint PD-L1. Anti-DEC205 scFv was anchored to the T. gondii surface either directly via glycosylphosphatidylinositol (GPI) or by fusion with the SAG1 protein. Both constructs were successfully expressed, but the binding results suggested that the anti-DEC-SAG1 scFv had more reliable functionality towards recombinant DEC protein and DEC205-expressing MutuDC cells. Two anti-PD-L1 scFv constructs were developed that differed in the localization of the HA tag. Both constructs were adequately expressed, but the localization of the HA tag determined the functionality by binding to PD-L1 protein. Co-incubation of T. gondii displaying anti-PD-L1 scFv with tumor cells expressing/displaying different levels of PD-L1 showed strong binding depending on the level of available biomarker. Neutralization assays confirmed that binding was due to the specific interaction between anti-PD-L1 scFv and its ligand. A mixed-cell assay showed that T. gondii expressing anti-PD-L1 scFv predominately targets the PD-L1-positive cells, with negligible off-target binding. The recombinant RH-PD-L1-C strain showed increased killing ability on PD-L1+ tumor cell lines compared to the parental strain. Moreover, a co-culture assay of target tumor cells and effector CD8+ T cells showed that our model could inhibit PD1/PD-L1 interaction and potentiate T-cell immune response. These findings highlight surface display of antibody fragments as a promising strategy of targeting replicative T. gondii strains while minimizing nonspecific binding.
    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
    一旦肺癌发生骨转移,治疗效率会大大降低。当前的主流治疗集中在抑制癌细胞生长和防止骨破坏。微波消融(MWA)已用于治疗骨肿瘤。然而,MWA可能会损害周围的正常组织。因此,开发纳米载体结合微波治疗骨转移可能是有益的。在这里,构建了一个微波响应的纳米平台(MgFe2O4@ZOL)。MgFe2O4@ZOLNPs释放Fe3+的货物,酸性肿瘤微环境(TME)中的Mg2+和唑来膦酸(ZOL)。Fe3+可以消耗细胞内谷胱甘肽(GSH)并催化H2O2生成•OH,导致化学动力学治疗(CDT)。此外,微波可以显着增强活性氧(ROS)的产生,从而使微波动态治疗(MDT)的有效实施。此外,Mg2+和ZOL促进成骨细胞分化。此外,MgFe2O4@ZOLNPs可以靶向并选择性加热肿瘤组织,并增强微波热疗(MTT)的效果。体外和体内实验都表明,协同靶向,GSH消耗增强CDT,MDT,选择性MTT具有显著的抗肿瘤疗效和骨修复作用。这种多模式联合治疗为肺癌患者骨转移的治疗提供了有希望的策略。
    Once bone metastasis occurs in lung cancer, the efficiency of treatment can be greatly reduced. Current mainstream treatments are focused on inhibiting cancer cell growth and preventing bone destruction. Microwave ablation (MWA) has been used to treat bone tumors. However, MWA may damage the surrounding normal tissues. Therefore, it could be beneficial to develop a nanocarrier combined with microwave to treat bone metastasis. Herein, a microwave-responsive nanoplatform (MgFe2O4@ZOL) was constructed. MgFe2O4@ZOL NPs release the cargos of Fe3+, Mg2+ and zoledronic acid (ZOL) in the acidic tumor microenvironment (TME). Fe3+ can deplete intracellular glutathione (GSH) and catalyze H2O2 to generate •OH, resulting in chemodynamic therapy (CDT). In addition, the microwave can significantly enhance the production of reactive oxygen species (ROS), thereby enabling the effective implementation of microwave dynamic therapy (MDT). Moreover, Mg2+ and ZOL promote osteoblast differentiation. In addition, MgFe2O4@ZOL NPs could target and selectively heat tumor tissue and enhance the effect of microwave thermal therapy (MTT). Both in vitro and in vivo experiments revealed that synergistic targeting, GSH depletion-enhanced CDT, MDT, and selective MTT exhibited significant antitumor efficacy and bone repair. This multimodal combination therapy provides a promising strategy for the treatment of bone metastasis in lung cancer 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
    虽然基于纳米平台的癌症治疗药物已经研究和调查了很多年,增强抗肿瘤疗效和减少毒副作用仍然是一个基本问题。
    我们利用铁(Fe2+)离子和端粒酶靶向发夹DNA结构之间的纳米粒子配位来封装阿霉素(DOX)和制造的Fe2+DNA@DOX纳米粒子(BDDFNPs)。这项工作研究了针对BDDFNPs的能力和生物分布的NIR荧光成像和药代动力学研究。体外和体内研究调查了纳米配方的毒性,成像,和协同治疗效果。
    增强的通透性和保留(EPR)效应和肿瘤靶向导致延长的血液循环时间和高的肿瘤积累。重要的是,根据正常细胞和肿瘤细胞端粒酶活性和铁依赖性的不同,BDDFNPs可以通过提高心肌细胞的抗氧化能力来降低DOX介导的心脏毒性。通过Fe2+介导的铁凋亡和β-连环蛋白/p53途径增强了协同治疗功效,并提高了肿瘤抑制率。
    基于HarpinDNA的纳米平台显示出延长的血液循环,通过端粒酶靶向的肿瘤药物积累,和协同治疗提高抗肿瘤药物疗效。我们的工作为未来的协同化疗提供了新的思路。
    UNASSIGNED: While nanoplatform-based cancer theranostics have been researched and investigated for many years, enhancing antitumor efficacy and reducing toxic side effects is still an essential problem.
    UNASSIGNED: We exploited nanoparticle coordination between ferric (Fe2+) ions and telomerase-targeting hairpin DNA structures to encapsulate doxorubicin (DOX) and fabricated Fe2+-DNA@DOX nanoparticles (BDDF NPs). This work studied the NIR fluorescence imaging and pharmacokinetic studies targeting the ability and biodistribution of BDDF NPs. In vitro and vivo studies investigated the nano formula\'s toxicity, imaging, and synergistic therapeutic effects.
    UNASSIGNED: The enhanced permeability and retention (EPR) effect and tumor targeting resulted in prolonged blood circulation times and high tumor accumulation. Significantly, BDDF NPs could reduce DOX-mediated cardiac toxicity by improving the antioxidation ability of cardiomyocytes based on the different telomerase activities and iron dependency in normal and tumor cells. The synergistic treatment efficacy is enhanced through Fe2+-mediated ferroptosis and the β-catenin/p53 pathway and improved the tumor inhibition rate.
    UNASSIGNED: Harpin DNA-based nanoplatforms demonstrated prolonged blood circulation, tumor drug accumulation via telomerase-targeting, and synergistic therapy to improve antitumor drug efficacy. Our work sheds new light on nanomaterials for future synergistic chemotherapy.
    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
    过度表达人表皮生长因子受体2(HER2)的乳腺癌预后不良。此外,可用的化学疗法由于选择性差而引起许多副作用。为了推进更有效和更安全的HER2阳性乳腺癌治疗,我们探索了药物递送技术和免疫治疗的融合。我们的研究导致了加载帕比司他和曲妥珠单抗抗体功能化的免疫囊体的设计,能够精确靶向过表达HER2的乳腺癌细胞。我们使用小角度X射线散射(SAXS)表征了立方体的纳米结构,低温透射电子显微镜(cryo-TEM),和动态光散射(DLS)。此外,我们通过傅里叶变换红外光谱(FTIR)和十二烷基硫酸钠聚丙烯酰胺凝胶电泳(SDS-PAGE)证实了免疫囊体上曲妥珠单抗抗体的完整性.此外,我们发现panobinostat负载的免疫囊体细胞毒性更大,以吸收依赖的方式,与代表健康细胞的细胞系(L929)相比,HER2阳性乳腺癌细胞系(SKBR3)。这些结果支持具有抗体的立方体的功能化增强了负载药物的有效性及其靶向HER2阳性乳腺癌细胞的选择性。
    Breast cancers that overexpress human epidermal growth factor receptor 2 (HER2) have poor prognosis. Moreover, available chemotherapies cause numerous side effects due to poor selectivity. To advance more effective and safer therapies for HER2-positive breast cancer, we explored the fusion of drug delivery technology and immunotherapy. Our research led to the design of immunocubosomes loaded with panobinostat and functionalized with trastuzumab antibodies, enabling precise targeting of breast cancer cells that overexpress HER2. We characterised the nanostructure of cubosomes using small-angle X-ray scattering (SAXS), cryo-transmission electron microscopy (cryo-TEM), and dynamic light scattering (DLS). Moreover, we confirmed the integrity of the trastuzumab antibodies on the immunocubosomes by Fourier-transform infrared spectroscopy (FTIR) and sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Additionally, we found that panobinostat-loaded immunocubosomes were more cytotoxic, and in an uptake-dependant manner, towards a HER2-positive breast cancer cell line (SKBR3) compared to a cell line representing healthy cells (L929). These results support that the functionalization of cubosomes with antibodies enhances both the effectiveness of the loaded drug and its selectivity for targeting HER2-positive breast cancer cells.
    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
    线粒体,最重要的细胞器之一,代表基础研究和生物医学应用的关键亚细胞靶标。尽管在各种生物应用的DNA纳米技术设计方面取得了重大进展,缺乏能够将线粒体靶向用于亚细胞分子成像和治疗的策略仍然是该领域的一个突出挑战.在这个Minireview中,我们总结了DNA纳米技术在线粒体靶向分子成像和肿瘤治疗中的设计和应用的最新进展。我们首先强调线粒体定位DNA纳米传感器的工程,用于对维持线粒体功能所必需的各种关键分子进行原位检测和成像,包括线粒体DNA和microRNA,酶,小分子,和金属离子。然后,我们汇编了DNA纳米技术的发展,用于线粒体靶向抗肿瘤治疗,包括模块化设计的DNA纳米设备,用于亚细胞递送治疗剂,和用于线粒体干扰的编程DNA组装。我们将重点阐明如何将DNA纳米生物技术设计为针对线粒体的各种生物医学应用的化学原理。最后,将讨论这一新兴领域的剩余挑战和未来方向,希望激发进一步开发用于线粒体学术和临床研究的先进DNA工具包。
    Mitochondria, one of the most important organelles, represent a crucial subcellular target for fundamental research and biomedical applications. Despite significant advances in the design of DNA nanotechnologies for a variety of bio-applications, the dearth of strategies that enable mitochondria targeting for subcellular molecular imaging and therapy remains an outstanding challenge in this field. In this Minireview, we summarize the recent progresses on the emerging design and application of DNA nanotechnology for mitochondria-targeted molecular imaging and tumor treatment. We first highlight the engineering of mitochondria-localized DNA nanosensors for in situ detection and imaging of diverse key molecules that are essential to maintain mitochondrial functions, including mitochondrial DNA and microRNA, enzymes, small molecules, and metal ions. Then, we compile the developments of DNA nanotechnologies for mitochondria-targeted anti-tumor therapy, including modularly designed DNA nanodevices for subcellular delivery of therapeutic agents, and programmed DNA assembly for mitochondrial interference. We will place an emphasis on clarification of the chemical principles of how DNA nanobiotechnology can be designed to target mitochondria for various biomedical applications. Finally, the remaining challenges and future directions in this emerging field will be discussed, hoping to inspire further development of advanced DNA toolkits for both academic and clinical research regarding mitochondria.
    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