Microneedle

微针
  • 文章类型: Journal Article
    巨噬细胞在糖尿病溃疡的愈合中起关键作用。葡萄糖水平的持续升高会损害巨噬细胞的胰岛素信号通路,导致功能失调的巨噬细胞难以从促炎(M1)过渡到修复(M2)状态。因此,通过胰岛素途径调节巨噬细胞炎症反应有望治疗糖尿病性溃疡。此外,生物膜的存在阻碍了药物的渗透,所产生的免疫抑制微环境加剧了促炎M1巨噬细胞的持续浸润。因此,我们设计了一系列可溶解的微针(表示为NPF@MN),装载有自组装纳米粒子,可以提供NPF纳米粒子,酸敏感性NPF释放原儿茶醛(PA),具有降血糖和胰岛素样作用,调节巨噬细胞极化为抗炎M2表型。此外,这项研究广泛研究了NPF@MN通过激活胰岛素信号通路加速糖尿病溃疡愈合的机制。通过RNA-seq和GSEA分析,我们发现途径相关因子如IR的表达减少,IRS-1、IRS-2和SHC。我们的工作提出了一种针对糖尿病溃疡胰岛素途径的创新治疗方法,并强调了其在临床管理中的转化潜力。
    Macrophages play a pivotal role in the healing of diabetic ulcers. The sustained elevation of glucose levels damages the insulin signaling pathway in macrophages, leading to dysfunctional macrophages that struggle to transition from pro-inflammatory (M1) to reparative (M2) states. Therefore, modulating macrophage inflammatory responses via the insulin pathway holds promise for diabetic ulcer treatment. Additionally, the presence of biofilm impedes drug penetration, and the resulting immunosuppressive microenvironment exacerbates the persistent infiltration of pro-inflammatory M1 macrophages. Therefore, we designed an array of dissolvable microneedle (denoted as NPF@MN) loaded with self-assembled nanoparticles that could deliver NPF nanoparticles, acid-sensitive NPF-releasing Protocatechualdehyde (PA) with hypoglycemic and insulin-like effects, regulating macrophage polarization to an anti-inflammatory M2 phenotype. Additionally, this study extensively examined the mechanism by which NPF@MN accelerates the healing of diabetic ulcers through the activation of the insulin signaling pathway. Through RNA-seq and GSEA analysis, we identified a reduction in the expression of pathway-related factors such as IR, IRS-1, IRS-2, and SHC. Our work presents an innovative therapeutic approach targeting the insulin pathway in diabetic ulcers and underscores its translational potential for clinical management.
    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
    一氧化碳(CO)已经成为一种有前途的治疗剂,然而,确保安全和精确的CO输送仍然具有挑战性。这里,我们报告了一种可移动的水凝胶形成微针(MN)反应器,用于通过光催化输送CO,重点是化学致敏。一经申请,MNs吸收的体液溶解泡腾剂,导致二氧化碳(CO2)的产生并触发化学疗法顺铂的释放。同时,在660nm的光照下,捕获在MNs内的光催化剂(PC)将CO2转化为CO。这些PC可以通过形成水凝胶的MNs去除,从而减轻与残留PC相关的潜在生物风险。体外和体内实验均表明,MN介导的CO递送通过抑制DNA修复显着提高了肿瘤对顺铂的敏感性,使用A375/CDDP黑色素瘤模型。这种可移动的光催化MN反应器提供安全和精确的CO局部输送,可能为CO或其联合疗法创造新的机会。
    Carbon monoxide (CO) has emerged as a promising therapeutic agent, yet ensuring safe and precise CO delivery remains challenging. Here, we report a removable hydrogel-forming microneedle (MN) reactor for CO delivery via photocatalysis, with an emphasis on chemosensitization. Upon application, body fluids absorbed by the MNs dissolve the effervescent agents, leading to the generation of carbon dioxide (CO2) and triggering the release of the chemotherapeutics cisplatin. Meanwhile, the photocatalysts (PCs) trapped within MNs convert CO2 to CO under 660 nm light irradiation. These PCs can be removed by hydrogel-forming MNs, thereby mitigating potential biological risks associated with residual PCs. Both in vitro and in vivo experiments showed that MN-mediated CO delivery significantly improved tumor sensitivity to cisplatin by suppressing DNA repair, using an A375/CDDP melanoma model. This removable photocatalysis MN reactor offers safe and precise local delivery of CO, potentially creating new opportunities for CO or its combination therapies.
    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
    伤口愈合是一个动态过程,涉及组织阶段的及时过渡。然而,感染的伤口通常会由于微生物超负荷而经历长时间的炎症。因此,解决不同愈合阶段的可行治疗需求是伤口管理的关键挑战。在这里,设计了一种新型的核-壳微针(CSMN)贴片,用于从德氏乳杆菌(LDEV)连续递送单宁酸-镁(TA-Mg)复合物和细胞外囊泡。一旦申请到受感染的网站,CSMN@TA-Mg/LDEV首先释放TA-Mg以抵消致病性超负荷并减少活性氧(ROS),帮助过渡到增殖阶段。随后,LDEV的持续释放增强角质形成细胞和成纤维细胞的活性,促进血管化,并调节胶原沉积。值得注意的是,微生物组成的动态跟踪表明,CSMN@TA-Mg/LDEV既可以抑制侵袭性病原体,又可以增加伤口部位的微生物多样性。功能分析进一步强调了CSMN@TA-Mg/LDEV促进伤口愈合和皮肤屏障恢复的潜力。此外,证实CSMN@TA-Mg/LDEV可以加速伤口闭合并改善鼠类感染伤口的恢复后皮肤质量。最后,这种创新的CSMN贴片为感染伤口提供了一种快速和高质量的替代治疗方法,并强调了微生物稳态的重要性。
    Wound healing is a dynamic process involving the timely transition of organized phases. However, infected wounds often experience prolonged inflammation due to microbial overload. Thus, addressing the viable treatment needs across different healing stages is a critical challenge in wound management. Herein, a novel core-shell microneedle (CSMN) patch is designed for the sequential delivery of tannic acid-magnesium (TA-Mg) complexes and extracellular vesicles from Lactobacillus druckerii (LDEVs). Upon application to infected sites, CSMN@TA-Mg/LDEV releases TA-Mg first to counteract pathogenic overload and reduce reactive oxygen species (ROS), aiding the transition to proliferative phase. Subsequently, the sustained release of LDEVs enhances the activities of keratinocytes and fibroblasts, promotes vascularization, and modulates the collagen deposition. Notably, dynamic track of microbial composition demonstrates that CSMN@TA-Mg/LDEV can both inhibit the aggressive pathogen and increase the microbial diversity at wound sites. Functional analysis further highlights the potential of CSMN@TA-Mg/LDEV in facilitating wound healing and skin barrier restoration. Moreover, it is confirmed that CSMN@TA-Mg/LDEV can accelerate wound closure and improve post-recovery skin quality in the murine infected wound. Conclusively, this innovative CSMN patch offers a rapid and high-quality alternative treatment for infected wounds and emphasizes the significance of microbial homeostasis.
    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
    透皮给药系统提供高生物利用度和良好的患者依从性,构成类风湿关节炎(RA)治疗局部给药的最佳方法。然而,角质层(SC)阻碍常规经皮药物递送系统的递送效率。微针(MNs)可以在SC内暂时产生微孔,通过绕过该屏障并增强透皮递送有效性来实现药物分布。值得注意的是,MN提供了通过皮肤递送药物的无痛方法,并且可以通过在经皮施用期间经由淋巴系统递送药物来直接调节免疫细胞中的炎症。然而,MN给药系统不适用于水溶性和稳定性较低的药物。此外,存在关于使用MN递送高细胞毒性药物的安全性的主要问题,考虑到这可能会导致分娩部位的局部药物浓度很高。虽然MN表现出一定程度的靶向递送至免疫和炎症环境,他们的瞄准效率仍然不理想。纳米制剂有可能通过改善药物靶向来显著解决RA治疗中MNs的局限性,溶解度,稳定性,和生物相容性。因此,这篇综述简要概述了这些优点,缺点,以及不同类型MNs治疗RA的机制。它特别着重于将纳米制剂与MNs结合用于RA治疗的应用和优势,并总结了纳米制剂与MNs结合在RA治疗领域中的当前发展趋势。为未来的进步和临床应用提供理论支持。
    A transdermal delivery system offers high bioavailability and favorable patient adherence, constituting an optimal approach for localized administration in rheumatoid arthritis (RA) treatment. However, the stratum corneum (SC) impedes the delivery efficiency of conventional transdermal drug delivery systems. Microneedles (MNs) can temporarily create micropores within the SC, enabling drug distribution via bypassing this barrier and enhancing transdermal delivery effectiveness. Notably, MNs provide a painless method of drug delivery through the skin and may directly modulate inflammation in immune cells by delivering drugs via the lymphatic system during transdermal administration. However, the MN delivery system is not suitable for drugs with low water solubility and stability. Additionally, major concerns exist regarding the safety of using MN delivery for highly cytotoxic drugs, given that it could result in high local drug concentration at the delivery site. While MNs exhibit some degree of targeted delivery to the immune and inflammatory environment, their targeting efficiency remains suboptimal. Nanoformulations have the potential to significantly address the limitations of MNs in RA treatment by improving drug targeting, solubility, stability, and biocompatibility. Therefore, this review provides a concise overview of the advantages, disadvantages, and mechanisms of different types of MNs for RA treatment. It specifically focuses on the application and advantages of combining nanoformulation with MNs for RA treatment and summarizes the current trends in the development of nanoformulations combined with MNs in the field of RA treatment, offering theoretical support for future advancements and clinical 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
    光老化对皮肤健康和衰老具有显著的重要性。紫外线(UV)照射导致细胞外基质(ECM)微环境的破坏,胶原蛋白的降解,和氧化应激的产生。传统的透明质酸(HA)表现出减弱的能力,刺激胶原蛋白再生,并受到其作为大分子的低渗透性的阻碍,最终导致光老化治疗效果受限。在这项研究中,HA/PX是通过用富含磺酸盐或富含磷脂酰胆碱的聚合物对HA进行功能改性来制备的,这可以补充ECM的损失并改善人成纤维细胞(HDFs)和UVB诱导的光老化的无毛小鼠模型的衰老。结果表明,HA/PX在延缓细胞衰老方面表现出优异的能力,促进胶原蛋白再生,与HA相比,抵抗活性氧(ROS)。此外,HA/PX在体内和体外均表现出良好的生物相容性,不会引起过敏反应或其他不良反应。我们还证明,通过微针阵列(MNs)经皮递送HA/PX可以显着减轻光老化裸鼠的皱纹和皮肤损伤,并通过增加表皮厚度来实现皮肤光老化的治疗,促进胶原蛋白沉积,减少氧化应激。因此,我们的研究为未来的抗衰老治疗策略提供了新的可能性.
    Photoaging holds remarkable importance for skin health and senescence. Ultraviolet (UV) irradiation results in the disruption of the extracellular matrix (ECM) microenvironment, the degradation of collagen, and the generation of oxidative stress. Traditional hyaluronic acid (HA) exhibits a diminished capacity to stimulate collagen regeneration, and hampered by its poor permeability as a macromolecule, ultimately resulting in constrained therapeutic outcomes for the treatment of photoaging. In this study, HA/PX was prepared by functional modification of HA with sulfonate-rich or phosphatidylcholine-rich polymers, which could complement the loss of ECM and ameliorate the senescence of human fibroblasts (HDFs) and hairless mouse models subjected to UVB-induced photoaging. The results indicate that HA/PX exhibits superior abilities in delaying cellular aging, promoting collagen regeneration, and resisting reactive oxygen species (ROS) compared to HA. Furthermore, HA/PX shows good biocompatibility both in vivo and in vitro, without causing allergic reactions or other adverse effects. We also demonstrated that the transdermal delivery of HA/PX via microneedle arrays (MNs) can significantly mitigate wrinkles and skin damage in photoaged nude mice, and achieve the treatment of skin photoaging by enhancing epidermal thickness, promoting collagen deposition, and reducing oxidative stress. Therefore, our research offers a novel possibility for future anti-aging therapeutic strategies.
    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
    肌腱干/祖细胞(TSPCs)对于肌腱修复至关重要,再生,和稳态。TSPCs的功能障碍,由于Janus激酶/信号转导和转录激活因子(JAK/STAT)信号通路的异常激活,有助于肌腱病。不幸的是,常规皮下注射靶向抑制JAK/STAT信号通路的有效性由于药物从损伤部位被动扩散而受到限制.在这里,设计了一种新型的聚γ-谷氨酸(γ-PGA)双倒钩微针(MN)路径,该路径负载了含有JAK/STAT抑制剂WP1066(MN-WP1066-NVs)的TSPC衍生的纳米囊泡(NVs),用于肌腱病的治疗。MN的双倒钩设计确保了对皮肤的牢固粘附,允许WP1066-NVs的持续和长期释放,促进增强的TSPCs自我更新,迁移,肌腱病的干性。体外和体内实验表明,γ-PGA贴片尖端的降解有助于WP1066-NVs在病变部位的逐渐释放。这种释放减轻了炎症,抑制细胞外基质降解,通过抑制JAK/STAT通路恢复正常肌腱组织结构。这些发现表明,多功能双倒钩MN贴片为肌腱病的治疗提供了一种新颖有效的治疗策略。
    Tendon stem/progenitor cells (TSPCs) are crucial for tendon repair, regeneration, and homeostasis. Dysfunction of TSPCs, due to aberrant activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway, contributes to tendinopathy. Unfortunately, the effectiveness of conventional subcutaneous injection targeting at suppressing JAK/STAT signaling pathway is limited due to the passive diffusion of drugs away from the injury site. Herein, a novel poly-gamma-glutamic acid (γ-PGA) dual-barb microneedle (MN) path loaded with TSPCs-derived nanovesicles (NVs) containing JAK/STAT inhibitor WP1066 (MN-WP1066-NVs) for tendinopathy treatment is designed. The dual-barb design of the MN ensures firm adhesion to the skin, allowing for sustained and prolonged release of WP1066-NVs, facilitating enhanced TSPCs self-renewal, migration, and stemness in tendinopathy. In vitro and in vivo experiments demonstrate that the degradation of γ-PGA patch tips facilitates the gradual release of WP1066-NVs at the lesion site. This release alleviates inflammation, suppresses extracellular matrix degradation, and restores normal tendon histological structure by inhibiting the JAK/STAT pathway. These findings suggest that the multifunctional dual-barb MN patch offers a novel and effective therapeutic strategy for tendinopathy 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
    本研究介绍了一种重复建模的新方法,以模拟人类复发性痛风发作的病理过程。这种方法解决了痛风大鼠模型中存在的不稳定性问题,提供了对人体骨骼系统造成的损害复发性痛风发作的更准确的表示。开发了一种包封秋水仙碱和艾拉莫德乙醇制剂的可溶性纳米针系统。该系统旨在调节炎症细胞因子并抑制破骨细胞活性,从而治疗与复发性痛风相关的炎性疼痛和骨损伤。此外,全面评估微针的外观,形态学,机械性能,和穿透能力证实了它们穿透角质层的有效性。溶解测试和皮肤刺激评估表明这些微针快速溶解而不刺激皮肤。体外渗透研究表明,与传统的局部应用相比,通过这些微针的透皮药物递送更有效并且导致更低的药物损失。在动物模型中进行的体内药效学评估显示,当两种类型的微针一起使用时,显著的镇痛和抗炎作用。进一步分析,包括X射线成像,苏木精和伊红(H&E)染色,Safranin-O/快速绿色染色,抗酒石酸酸性磷酸酶染色,和破骨细胞的定量,证实了微针组合的骨保护作用。总之,这项研究的结果强调了这种新的治疗方法在复发性痛风的临床应用中的潜力。
    This study introduces a novel approach of repetitive modeling to simulate the pathological process of recurrent gout attacks in humans. This methodology addresses the instability issues present in rat models of gout, providing a more accurate representation of the damage recurrent gout episodes inflict on human skeletal systems. A soluble nanoneedle system encapsulating colchicine and iguratimod ethosomal formulations was developed. This system aims to modulate inflammatory cytokines and inhibit osteoclast activity, thereby treating inflammatory pain and bone damage associated with recurrent gout. Additionally, a comprehensive evaluation of the microneedles\' appearance, morphology, mechanical properties, and penetration capability confirmed their effectiveness in penetrating the stratum corneum. Dissolution tests and skin irritation assessments demonstrated that these microneedles dissolve rapidly without irritating the skin. In vitro permeation studies indicated that transdermal drug delivery via these microneedles is more efficient and incurs lower drug loss compared to traditional topical applications. In vivo pharmacodynamic assessments conducted in animal models revealed significant analgesic and anti-inflammatory effects when both types of microneedles were used together. Further analyses, including X-ray imaging, hematoxylin and eosin (H&E) staining, Safranin-O/fast green staining, tartrate-resistant acid phosphatase staining, and quantification of osteoclasts, confirmed the bone-protective effects of the microneedle combination. In conclusion, the findings of this research underscore the potential of this novel therapeutic approach for clinical application in the treatment of recurrent gout.
    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
    由于细胞增殖和血管生成不足而导致的伤口愈合受损是全世界患者的重大生理和心理负担。用于伤口修复的高剂量的外源性生长因子(GF)的治疗性递送是不理想的,因为GF在蛋白水解伤口环境中具有差的稳定性。这里,我们提出了一种两阶段策略,使用基于硫糖铝的生物活性微针(SUC-MN)递送白细胞介素-4(IL-4)以加速伤口愈合.在第一阶段,SUC-MN通过经由JAK-STAT途径更有效地重编程促再生M2样巨噬细胞以增加内源性GF产生来协同地增强IL-4的作用。在第二阶段,硫糖铝与GFs结合,并在空间上不利于蛋白酶降解,以增加GFs的生物利用度。与各自未处理的对照相比,IL-4/SUC-MN技术在糖尿病小鼠伤口和猪伤口中加速伤口愈合56.6%和46.5%。总的来说,我们的研究结果强调了创新使用分子模拟来识别生物活性成分,并将其掺入微针中,通过多种协同机制促进伤口愈合.
    Impaired wound healing due to insufficient cell proliferation and angiogenesis is a significant physical and psychological burden to patients worldwide. Therapeutic delivery of exogenous growth factors (GFs) at high doses for wound repair is non-ideal as GFs have poor stability in proteolytic wound environments. Here, we present a two-stage strategy using bioactive sucralfate-based microneedle (SUC-MN) for delivering interleukin-4 (IL-4) to accelerate wound healing. In the first stage, SUC-MN synergistically enhanced the effect of IL-4 through more potent reprogramming of pro-regenerative M2-like macrophages via the JAK-STAT pathway to increase endogenous GF production. In the second stage, sucralfate binds to GFs and sterically disfavors protease degradation to increase bioavailability of GFs. The IL-4/SUC-MN technology accelerated wound healing by 56.6 % and 46.5 % in diabetic mice wounds and porcine wounds compared to their respective untreated controls. Overall, our findings highlight the innovative use of molecular simulations to identify bioactive ingredients and their incorporation into microneedles for promoting wound healing through multiple synergistic mechanisms.
    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
    口腔溃疡,口腔粘膜表面的浅表病变,发病率很高,其主要症状包括局部疼痛和糜烂。脂多糖(LPS)预处理的骨髓间充质干细胞及其分泌的外泌体(LPS-pre-Exos)已显示可促进各种炎症和伤口的恢复。然而,缺乏记录LPS-pre-Exos作为口腔粘膜样疾病治疗干预措施的研究。在这项研究中,我们制备了由LPS-pre-Exos和位于尖端和基部的沸石咪唑酯框架-8(ZIF-8)组成的丝素蛋白微针(MN)贴片,分别,并使用这种MN贴片治疗口腔溃疡。插入口腔粘膜后,观察到持续的LPS-Exos释放,促进巨噬细胞极化和组织愈合。此外,MN贴片中的ZIF-8框架促进了Zn2+的控释,通过协同效应证明了有效的抗菌性能。体外实验结果表明,丝素蛋白MN贴片可持续释放LPS-pre-Exos和Zn2+超过7天。因此,LPS-pre-Exos和ZIF-8负载丝素蛋白MN贴片表现出良好的抗炎和抗菌性能,促进口腔溃疡愈合,并显示出良好的组织相容性。因此,它可能是促进口腔溃疡愈合的潜在有价值的策略。
    Oral ulcers, superficial lesions on the surface of the oral mucosa, have a high incidence rate, and their main symptoms include local pain and erosion. Lipopolysaccharide (LPS)-preconditioned bone marrow mesenchymal stem cells and their secreted exosomes (LPS-pre-Exos) have been shown to promote recovery in various inflammatory conditions and wounds. However, studies documenting LPS-pre-Exos as a therapeutic intervention for oral mucosal-like diseases are lacking. In this study, we prepared a silk fibroin microneedle (MN) patch consisting of LPS-pre-Exos and zeolitic imidazolate framework-8 (ZIF-8) that localized at the tip and base, respectively, and used this MN patch for oral ulcer treatment. Upon insertion into the oral mucosa, continuous LPS-pre-Exos release was observed, which promoted macrophage polarization and tissue healing. Additionally, the ZIF-8 framework in the MN patch facilitated the controlled release of Zn2+, which demonstrated potent antimicrobial properties via synergistic effects. The in vitro experimental results showed that the silk fibroin MN patch can continuously release LPS-pre-Exos and Zn2+ for more than 7 days. Thus, the LPS-pre-Exos and ZIF-8-loaded silk fibroin MN patch exhibited good anti-inflammatory and antibacterial properties, promoting oral ulcer healing, and showed good histocompatibility. Hence, it may represent a potentially valuable strategy for facilitating oral ulcer healing.
    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
    胃幽门螺杆菌感染是胃黏膜出血的主要原因。由于存在胃粘膜层,通过口服治疗消除胃幽门螺杆菌仍然很困难,它作为口服药物的物理屏障。在这项研究中,用于口服给药的磁性导航微针药物递送平台(MNsD),具有差分双模药物释放速率,旨在实现快速的胃止血并克服胃屏障,以长期抑制胃中的幽门螺杆菌。MNs-D是通过合理地加载载体衬底来创建的,由具有可变溶解度的丝素蛋白组成,抗生素和止血剂.体外实验表明,MNs-D可以持久地根除刺激的胃液中的幽门螺杆菌,并具有持久的药物释放(24小时内占79%),并迅速建立止血并立即释放药物(60s内占92%)。最重要的是,体内研究表明,MNs-D通过在磁导航下插入GML,克服了传统口服给药疗法中不稳定的胃粘膜屏障,导致持续的抗生素释放,长期持续的幽门螺杆菌感染(99%)。对于针对胃幽门螺杆菌感染的差异双模式药物释放,这项研究可能首先检查了口服磁性导航微针的效果。
    The Helicobacter pylori infection in the stomach is the key reason for gastric mucosal bleeding. Eliminating gastric Helicobacter pylori by oral treatment remains difficult due to the presence of the gastric mucosal layer, which acts as a physical barrier to drugs via oral administration. In this study, a magnetic-navigable microneedle drug delivery platform (MNsD) for oral administration, featuring differential dual-mode drug release rate, was designed to fulfil rapid gastric hemostasis and overcome the gastric barriers for long-lasting Helicobacter pylori inhibition in stomach. MNs-D was created by rationally loading the carrier substrate, which was composed of silk fibroin with variable solubility, with antibiotics and hemostats. In vitro experiments showed MNs-D may sustainably eradicate Helicobacter pylori in stimulated gastric juices with long-lasting drug release (79 % in 24 h) and quickly establish hemostasis with instant drug release (92 % within 60 s). Most importantly, in vivo studies demonstrated MNs-D overcame the unsettling gastric mucosal barrier in traditional therapies of oral administration by insertion into the GML under magnetic navigation, resulting in sustained antibiotic release for long-lasting Helicobacter pylori eradiation (99 %). For differential dual-mode medication release against gastric Helicobacter pylori infections, this study may have firstly examined the effects of magnetic navigated microneedles administered orally.
    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