首页 > 最新文献

Journal of Nanobiotechnology最新文献

英文 中文
A novel spherical GelMA-HAMA hydrogel encapsulating APET×2 polypeptide and CFIm25-targeting sgRNA for immune microenvironment modulation and nucleus pulposus regeneration in intervertebral discs 包裹 APET×2 多肽和 CFIm25 靶向 sgRNA 的新型球形 GelMA-HAMA 水凝胶,用于调节免疫微环境和椎间盘髓核再生
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-12 DOI: 10.1186/s12951-024-02783-z
Xiao-Jun Yu, Yuan-Ting Zhao, Haimiti Abudouaini, Peng Zou, Tian-Qi Li, Xiao-Fan Bai, Shan-Xi Wang, Jian-Bin Guan, Meng-wei Li, Xiao-dong Wang, Ying-guang Wang, Ding-Jun Hao
Single-cell transcriptomics and high-throughput transcriptomics were used to screen factors significantly correlated with intervertebral disc degeneration (IDD). Expression changes of CFIm25 were determined via RT-qPCR and Western blot. NP cells were isolated from mouse intervertebral discs and induced to degrade with TNF-α and IL-1β. CFIm25 was knocked out using CRISPR-Cas9, and CFIm25 knockout and overexpressing nucleus pulposus (NP) cell lines were generated through lentiviral transfection. Proteoglycan expression, protein expression, inflammatory factor expression, cell viability, proliferation, migration, gene expression, and protein expression were analyzed using various assays (alcian blue staining, immunofluorescence, ELISA, CCK-8, EDU labeling, transwell migration, scratch assay, RT-qPCR, Western blot). The GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA was designed, and its effects on NP regeneration were assessed through in vitro and mouse model experiments. The progression of IDD in mice was evaluated using X-ray, H&E staining, and Safranin O-Fast Green staining. Immunohistochemistry was performed to determine protein expression in NP tissue. Proteomic analysis combined with in vitro and in vivo experiments was conducted to elucidate the mechanisms of hydrogel action. CFIm25 was upregulated in IDD NP tissue and significantly correlated with disease progression. Inhibition of CFIm25 improved NP cell degeneration, enhanced cell proliferation, and migration. The hydrogel effectively knocked down CFIm25 expression, improved NP cell degeneration, promoted cell proliferation and migration, and mitigated IDD progression in a mouse model. The hydrogel inhibited inflammatory factor expression (IL-6, iNOS, IL-1β, TNF-α) by targeting the p38/NF-κB signaling pathway, increased collagen COLII and proteoglycan Aggrecan expression, and suppressed NP degeneration-related factors (COX-2, MMP-3). The study highlighted the crucial role of CFIm25 in IDD and introduced a promising therapeutic strategy using a porous spherical GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA. This innovative approach offers new possibilities for treating degenerated intervertebral discs.
利用单细胞转录组学和高通量转录组学筛选与椎间盘变性(IDD)显著相关的因素。通过 RT-qPCR 和 Western 印迹测定了 CFIm25 的表达变化。从小鼠椎间盘中分离出NP细胞,用TNF-α和IL-1β诱导其退化。使用CRISPR-Cas9敲除CFIm25,并通过慢病毒转染产生CFIm25敲除和过表达的髓核细胞系。通过各种检测方法(藻蓝染色、免疫荧光、ELISA、CCK-8、EDU 标记、经孔迁移、划痕试验、RT-qPCR、Western 印迹)对蛋白多糖表达、蛋白质表达、炎症因子表达、细胞活力、增殖、迁移、基因表达和蛋白质表达进行了分析。设计了负载 APET×2 多肽和 sgRNA 的 GelMA-HAMA 水凝胶,并通过体外实验和小鼠模型实验评估了其对 NP 再生的影响。使用 X 光、H&E 染色和 Safranin O-Fast Green 染色评估了小鼠 IDD 的进展情况。免疫组织化学测定了 NP 组织中的蛋白质表达。蛋白质组分析与体外和体内实验相结合,阐明了水凝胶的作用机制。CFIm25 在 IDD NP 组织中上调,并与疾病进展显著相关。抑制 CFIm25 可改善 NP 细胞变性,增强细胞增殖和迁移。在小鼠模型中,水凝胶能有效抑制 CFIm25 的表达,改善 NP 细胞变性,促进细胞增殖和迁移,缓解 IDD 的进展。水凝胶通过靶向 p38/NF-κB 信号通路抑制了炎症因子(IL-6、iNOS、IL-1β、TNF-α)的表达,增加了胶原 COLII 和蛋白多糖 Aggrecan 的表达,抑制了 NP 退化相关因子(COX-2、MMP-3)。该研究强调了 CFIm25 在 IDD 中的关键作用,并介绍了一种使用多孔球形 GelMA-HAMA 水凝胶加载 APET×2 多肽和 sgRNA 的有前景的治疗策略。这种创新方法为治疗退化的椎间盘提供了新的可能性。
{"title":"A novel spherical GelMA-HAMA hydrogel encapsulating APET×2 polypeptide and CFIm25-targeting sgRNA for immune microenvironment modulation and nucleus pulposus regeneration in intervertebral discs","authors":"Xiao-Jun Yu, Yuan-Ting Zhao, Haimiti Abudouaini, Peng Zou, Tian-Qi Li, Xiao-Fan Bai, Shan-Xi Wang, Jian-Bin Guan, Meng-wei Li, Xiao-dong Wang, Ying-guang Wang, Ding-Jun Hao","doi":"10.1186/s12951-024-02783-z","DOIUrl":"https://doi.org/10.1186/s12951-024-02783-z","url":null,"abstract":"Single-cell transcriptomics and high-throughput transcriptomics were used to screen factors significantly correlated with intervertebral disc degeneration (IDD). Expression changes of CFIm25 were determined via RT-qPCR and Western blot. NP cells were isolated from mouse intervertebral discs and induced to degrade with TNF-α and IL-1β. CFIm25 was knocked out using CRISPR-Cas9, and CFIm25 knockout and overexpressing nucleus pulposus (NP) cell lines were generated through lentiviral transfection. Proteoglycan expression, protein expression, inflammatory factor expression, cell viability, proliferation, migration, gene expression, and protein expression were analyzed using various assays (alcian blue staining, immunofluorescence, ELISA, CCK-8, EDU labeling, transwell migration, scratch assay, RT-qPCR, Western blot). The GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA was designed, and its effects on NP regeneration were assessed through in vitro and mouse model experiments. The progression of IDD in mice was evaluated using X-ray, H&E staining, and Safranin O-Fast Green staining. Immunohistochemistry was performed to determine protein expression in NP tissue. Proteomic analysis combined with in vitro and in vivo experiments was conducted to elucidate the mechanisms of hydrogel action. CFIm25 was upregulated in IDD NP tissue and significantly correlated with disease progression. Inhibition of CFIm25 improved NP cell degeneration, enhanced cell proliferation, and migration. The hydrogel effectively knocked down CFIm25 expression, improved NP cell degeneration, promoted cell proliferation and migration, and mitigated IDD progression in a mouse model. The hydrogel inhibited inflammatory factor expression (IL-6, iNOS, IL-1β, TNF-α) by targeting the p38/NF-κB signaling pathway, increased collagen COLII and proteoglycan Aggrecan expression, and suppressed NP degeneration-related factors (COX-2, MMP-3). The study highlighted the crucial role of CFIm25 in IDD and introduced a promising therapeutic strategy using a porous spherical GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA. This innovative approach offers new possibilities for treating degenerated intervertebral discs. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224937","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Preparation and effects of functionalized liposomes targeting breast cancer tumors using chemotherapy, phototherapy, and immunotherapy 利用化疗、光疗和免疫疗法靶向乳腺癌肿瘤的功能化脂质体的制备和效果
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-12 DOI: 10.1186/s12951-024-02838-1
Bowen Zeng, Lina Pian, Yanhong Liu, Shuangqing Wang, Nuoya Wang, Chao Liu, Hao Wu, Hongshuang Wan, Liqing Chen, Wei Huang, Zhonggao Gao, Xuezhe Yin, Mingji Jin
Breast cancer therapy has significantly advanced by targeting the programmed cell death-ligand 1/programmed cell death-1 (PD-L1/PD-1) pathway. BMS-202 (a smallmolecule PD-L1 inhibitor) induces PD-L1 dimerization to block PD-1/PD-L1 interactions, allowing the T-cell-mediated immune response to kill tumor cells. However, immunotherapy alone has limited effects. Clinically approved photodynamic therapy (PDT) activates immunity and selectively targets malignant cells. However, PDT aggravates hypoxia, which may compromise its therapeutic efficacy and promote tumor metastasis. We designed a tumor-specific delivery nanoplatform of liposomes that encapsulate the hypoxia-sensitive antitumor drug tirapazamine (TPZ) and the small-molecule immunosuppressant BMS. New indocyanine green (IR820)-loaded polyethylenimine-folic acid (PEI-FA) was complexed with TPZ and BMS-loaded liposomes via electrostatic interactions to form lipid nanocomposites. This nanoplatform can be triggered by near-infrared irradiation to induce PDT, resulting in a hypoxic tumor environment and activation of the prodrug TPZ to achieve efficient chemotherapy. The in vitro and in vivo studies demonstrated excellent combined PDT, chemotherapy, and immunotherapy effects on the regression of distant tumors and lung metastases, providing a reference method for the preparation of targeted agents for treating breast cancer.
针对程序性细胞死亡配体1/程序性细胞死亡-1(PD-L1/PD-1)途径的乳腺癌治疗取得了重大进展。BMS-202(一种小分子 PD-L1 抑制剂)可诱导 PD-L1 二聚化,从而阻断 PD-1/PD-L1 的相互作用,使 T 细胞介导的免疫反应能够杀死肿瘤细胞。然而,单纯的免疫疗法效果有限。临床批准的光动力疗法(PDT)可激活免疫,并选择性地靶向恶性细胞。然而,光动力疗法会加重缺氧,从而影响疗效并促进肿瘤转移。我们设计了一种肿瘤特异性递送纳米脂质体平台,其中封装了对缺氧敏感的抗肿瘤药物替拉帕胺(TPZ)和小分子免疫抑制剂 BMS。新型吲哚菁绿(IR820)负载的聚亚乙基亚胺叶酸(PEI-FA)通过静电作用与TPZ和BMS负载的脂质体复合,形成脂质纳米复合体。该纳米平台可通过近红外照射诱导光导放疗,形成缺氧的肿瘤环境并激活原药 TPZ,从而实现高效化疗。体外和体内研究表明,PDT、化疗和免疫疗法联合使用对远处肿瘤和肺转移灶的消退有很好的效果,为制备治疗乳腺癌的靶向药物提供了一种参考方法。
{"title":"Preparation and effects of functionalized liposomes targeting breast cancer tumors using chemotherapy, phototherapy, and immunotherapy","authors":"Bowen Zeng, Lina Pian, Yanhong Liu, Shuangqing Wang, Nuoya Wang, Chao Liu, Hao Wu, Hongshuang Wan, Liqing Chen, Wei Huang, Zhonggao Gao, Xuezhe Yin, Mingji Jin","doi":"10.1186/s12951-024-02838-1","DOIUrl":"https://doi.org/10.1186/s12951-024-02838-1","url":null,"abstract":"Breast cancer therapy has significantly advanced by targeting the programmed cell death-ligand 1/programmed cell death-1 (PD-L1/PD-1) pathway. BMS-202 (a smallmolecule PD-L1 inhibitor) induces PD-L1 dimerization to block PD-1/PD-L1 interactions, allowing the T-cell-mediated immune response to kill tumor cells. However, immunotherapy alone has limited effects. Clinically approved photodynamic therapy (PDT) activates immunity and selectively targets malignant cells. However, PDT aggravates hypoxia, which may compromise its therapeutic efficacy and promote tumor metastasis. We designed a tumor-specific delivery nanoplatform of liposomes that encapsulate the hypoxia-sensitive antitumor drug tirapazamine (TPZ) and the small-molecule immunosuppressant BMS. New indocyanine green (IR820)-loaded polyethylenimine-folic acid (PEI-FA) was complexed with TPZ and BMS-loaded liposomes via electrostatic interactions to form lipid nanocomposites. This nanoplatform can be triggered by near-infrared irradiation to induce PDT, resulting in a hypoxic tumor environment and activation of the prodrug TPZ to achieve efficient chemotherapy. The in vitro and in vivo studies demonstrated excellent combined PDT, chemotherapy, and immunotherapy effects on the regression of distant tumors and lung metastases, providing a reference method for the preparation of targeted agents for treating breast cancer. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ACYP2 functions as an innovative nano-therapeutic target to impede the progression of hepatocellular carcinoma by inhibiting the activity of TERT and the KCNN4/ERK pathway ACYP2 通过抑制 TERT 和 KCNN4/ERK 通路的活性,成为阻碍肝细胞癌进展的创新纳米治疗靶点
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-12 DOI: 10.1186/s12951-024-02827-4
Yixuan Wu, Hongyi Bao, Jinran Wu, Bairong Chen, Jing Xu, Kangfeng Jin, Lin Chen, Guang Zhu, Feng Wang
An increasing body of evidence suggests that acylphosphatase-2 (ACYP2) polymorphisms are correlated with an increased susceptibility to a range of malignancies. Nevertheless, its potential functions, molecular mechanisms in hepatocellular carcinoma (HCC) and whether it can be act as a therapeutic target remain uninvestigated. Herein, ACYP2 was found to be lowly expressed in HCC and was negatively correlated with tumor size, tumor differentiation, microvascular invasion and the prognosis of HCC patients. Functional investigations revealed that overexpression of ACYP2 inhibited the proliferation and metastasis of HCC cells while promoting apoptosis; knockdown of ACYP2 had the exact opposite effect. Additionally, it was observed that ACYP2 was distributed in both the cytoplasm and nucleus of HCC cells. According to the mechanistic studies, the expression of potassium calcium-activated channel subfamily N member 4 (KCNN4) was negatively regulated by cytoplasmic ACYP2, resulting in the inhibition of K+ outflow and subsequent inactivation of the ERK pathway, which impeded the growth and metastasis of HCC. Furthermore, the activity of telomerase reverse transcriptase (TERT) was inhibited by nuclear ACYP2, leading to the reduction in length of telomeres and consequent reversal of HCC cell immortalization. Additionally, a novel targeted nanotherapy strategy was developed wherein the pcDNA-ACYP2 vector was encapsulated within polyetherimide nanoparticles (PEI/NPs), which were subsequently coated with HCC cell membranes (namely pcDNA/PEI/NPs@M). Safety and targeting characteristics abound for these nanocomposites, in both subcutaneous graft tumor models and orthotopic mouse models, they inhibited the progression of HCC by impeding TERT activity and the KCNN4/ERK pathway. In conclusion, our research identifies novel molecular mechanisms involving cytoplasmic and nuclear ACYP2 that inhibit the progression of HCC. Moreover, pcDNA/PEI/NPs@M represents a targeted therapeutic strategy for HCC that holds great promising.
越来越多的证据表明,酰基磷酸酶-2(ACYP2)多态性与一系列恶性肿瘤的易感性增加有关。然而,它在肝细胞癌(HCC)中的潜在功能、分子机制以及是否可作为治疗靶点仍有待研究。本文发现 ACYP2 在 HCC 中低表达,并与 HCC 患者的肿瘤大小、肿瘤分化、微血管侵犯和预后呈负相关。功能研究发现,过表达 ACYP2 可抑制 HCC 细胞的增殖和转移,同时促进细胞凋亡;而敲除 ACYP2 的效果恰恰相反。此外,还观察到 ACYP2 同时分布于 HCC 细胞的细胞质和细胞核中。机理研究发现,钾钙激活通道 N 亚家族成员 4(KCNN4)的表达受细胞质中 ACYP2 的负调控,导致 K+ 外流受抑制,ERK 通路随之失活,从而阻碍了 HCC 的生长和转移。此外,核 ACYP2 还抑制了端粒酶逆转录酶(TERT)的活性,导致端粒长度缩短,从而逆转了 HCC 细胞的永生化。此外,还开发了一种新的靶向纳米疗法策略,将 pcDNA-ACYP2 载体封装在聚醚酰亚胺纳米颗粒(PEI/NPs)中,然后在其上包覆 HCC 细胞膜(即 pcDNA/PEI/NPs@M)。这些纳米复合材料具有安全性和靶向性,在皮下移植肿瘤模型和小鼠正位模型中,它们都能通过抑制TERT活性和KCNN4/ERK通路来抑制HCC的进展。总之,我们的研究发现了涉及细胞质和细胞核 ACYP2 的抑制 HCC 进展的新型分子机制。此外,pcDNA/PEI/NPs@M代表了一种治疗HCC的靶向治疗策略,具有广阔的前景。
{"title":"ACYP2 functions as an innovative nano-therapeutic target to impede the progression of hepatocellular carcinoma by inhibiting the activity of TERT and the KCNN4/ERK pathway","authors":"Yixuan Wu, Hongyi Bao, Jinran Wu, Bairong Chen, Jing Xu, Kangfeng Jin, Lin Chen, Guang Zhu, Feng Wang","doi":"10.1186/s12951-024-02827-4","DOIUrl":"https://doi.org/10.1186/s12951-024-02827-4","url":null,"abstract":"An increasing body of evidence suggests that acylphosphatase-2 (ACYP2) polymorphisms are correlated with an increased susceptibility to a range of malignancies. Nevertheless, its potential functions, molecular mechanisms in hepatocellular carcinoma (HCC) and whether it can be act as a therapeutic target remain uninvestigated. Herein, ACYP2 was found to be lowly expressed in HCC and was negatively correlated with tumor size, tumor differentiation, microvascular invasion and the prognosis of HCC patients. Functional investigations revealed that overexpression of ACYP2 inhibited the proliferation and metastasis of HCC cells while promoting apoptosis; knockdown of ACYP2 had the exact opposite effect. Additionally, it was observed that ACYP2 was distributed in both the cytoplasm and nucleus of HCC cells. According to the mechanistic studies, the expression of potassium calcium-activated channel subfamily N member 4 (KCNN4) was negatively regulated by cytoplasmic ACYP2, resulting in the inhibition of K+ outflow and subsequent inactivation of the ERK pathway, which impeded the growth and metastasis of HCC. Furthermore, the activity of telomerase reverse transcriptase (TERT) was inhibited by nuclear ACYP2, leading to the reduction in length of telomeres and consequent reversal of HCC cell immortalization. Additionally, a novel targeted nanotherapy strategy was developed wherein the pcDNA-ACYP2 vector was encapsulated within polyetherimide nanoparticles (PEI/NPs), which were subsequently coated with HCC cell membranes (namely pcDNA/PEI/NPs@M). Safety and targeting characteristics abound for these nanocomposites, in both subcutaneous graft tumor models and orthotopic mouse models, they inhibited the progression of HCC by impeding TERT activity and the KCNN4/ERK pathway. In conclusion, our research identifies novel molecular mechanisms involving cytoplasmic and nuclear ACYP2 that inhibit the progression of HCC. Moreover, pcDNA/PEI/NPs@M represents a targeted therapeutic strategy for HCC that holds great promising. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224936","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Curcumin nanoparticles in heat stroke management 姜黄素纳米颗粒在中暑治疗中的应用
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-12 DOI: 10.1186/s12951-024-02771-3
Fei Guo, Yizhan Wu, Jiangwei Liu
The exacerbation of extreme high-temperature events due to global climate change poses a significant challenge to public health, particularly impacting the central nervous system through heat stroke. This study aims to develop Poly(amidoamine) (PAMAM) nanoparticles loaded with curcumin (PAMAM@Cur) to enhance its therapeutic efficacy in hypothalamic neural damage in a heat stroke model and explore its potential mechanisms. Curcumin (Cur) was encapsulated into PAMAM nanoparticles through a hydrophobic interaction method, and various techniques were employed to characterize their physicochemical properties. A heat stroke mouse model was established to monitor body temperature and serum biochemical parameters, conduct behavioral assessments, histological examinations, and biochemical analyses. Transcriptomic and proteomic analyses were performed to investigate the therapeutic mechanisms of PAMAM@Cur, validated in an N2a cell model. PAMAM@Cur demonstrated good stability, photostability, cell compatibility, significant blood–brain barrier (BBB) penetration capability, and effective accumulation in the brain. PAMAM@Cur markedly improved behavioral performance and neural cell structural integrity in heat stroke mice, alleviated inflammatory responses, with superior therapeutic effects compared to Cur or PAMAM alone. Multi-omics analysis revealed that PAMAM@Cur regulated antioxidant defense genes and iron death-related genes, particularly upregulating the PCBP2 protein, stabilizing SLC7A11 and GPX4 mRNA, and reducing iron-dependent cell death. By enhancing the drug delivery properties of Cur and modulating molecular pathways relevant to disease treatment, PAMAM@Cur significantly enhances the therapeutic effects against hypothalamic neural damage induced by heat stroke, showcasing the potential of nanotechnology in improving traditional drug efficacy and providing new strategies for future clinical applications. This study highlights the outlook of nanotechnology in treating neurological disorders caused by heat stroke, offering a novel therapeutic approach with potential clinical applications.
全球气候变化导致极端高温事件加剧,给公共卫生带来了重大挑战,尤其是中暑对中枢神经系统的影响。本研究旨在开发负载姜黄素(PAMAM@Cur)的聚酰胺胺(PAMAM)纳米粒子,以增强其在中暑模型中对下丘脑神经损伤的疗效,并探索其潜在机制。通过疏水相互作用方法将姜黄素(Cur)封装到 PAMAM 纳米颗粒中,并采用多种技术表征其理化性质。建立中暑小鼠模型,监测体温和血清生化指标,进行行为评估、组织学检查和生化分析。为了研究 PAMAM@Cur 的治疗机制,还进行了转录组和蛋白质组分析,并在 N2a 细胞模型中进行了验证。PAMAM@Cur表现出良好的稳定性、光稳定性、细胞相容性、显著的血脑屏障(BBB)穿透能力以及在大脑中的有效蓄积。PAMAM@Cur 显著改善了中暑小鼠的行为表现和神经细胞结构的完整性,缓解了炎症反应,治疗效果优于单独使用 Cur 或 PAMAM。多组学分析表明,PAMAM@Cur能调节抗氧化防御基因和铁死亡相关基因,特别是上调PCBP2蛋白,稳定SLC7A11和GPX4 mRNA,减少铁依赖性细胞死亡。PAMAM@Cur 通过增强 Cur 的给药特性和调节与疾病治疗相关的分子通路,显著提高了对中暑引起的下丘脑神经损伤的治疗效果,展示了纳米技术在改善传统药物疗效方面的潜力,并为未来的临床应用提供了新的策略。这项研究强调了纳米技术在治疗中暑引起的神经系统疾病方面的前景,提供了一种具有潜在临床应用价值的新型治疗方法。
{"title":"Curcumin nanoparticles in heat stroke management","authors":"Fei Guo, Yizhan Wu, Jiangwei Liu","doi":"10.1186/s12951-024-02771-3","DOIUrl":"https://doi.org/10.1186/s12951-024-02771-3","url":null,"abstract":"The exacerbation of extreme high-temperature events due to global climate change poses a significant challenge to public health, particularly impacting the central nervous system through heat stroke. This study aims to develop Poly(amidoamine) (PAMAM) nanoparticles loaded with curcumin (PAMAM@Cur) to enhance its therapeutic efficacy in hypothalamic neural damage in a heat stroke model and explore its potential mechanisms. Curcumin (Cur) was encapsulated into PAMAM nanoparticles through a hydrophobic interaction method, and various techniques were employed to characterize their physicochemical properties. A heat stroke mouse model was established to monitor body temperature and serum biochemical parameters, conduct behavioral assessments, histological examinations, and biochemical analyses. Transcriptomic and proteomic analyses were performed to investigate the therapeutic mechanisms of PAMAM@Cur, validated in an N2a cell model. PAMAM@Cur demonstrated good stability, photostability, cell compatibility, significant blood–brain barrier (BBB) penetration capability, and effective accumulation in the brain. PAMAM@Cur markedly improved behavioral performance and neural cell structural integrity in heat stroke mice, alleviated inflammatory responses, with superior therapeutic effects compared to Cur or PAMAM alone. Multi-omics analysis revealed that PAMAM@Cur regulated antioxidant defense genes and iron death-related genes, particularly upregulating the PCBP2 protein, stabilizing SLC7A11 and GPX4 mRNA, and reducing iron-dependent cell death. By enhancing the drug delivery properties of Cur and modulating molecular pathways relevant to disease treatment, PAMAM@Cur significantly enhances the therapeutic effects against hypothalamic neural damage induced by heat stroke, showcasing the potential of nanotechnology in improving traditional drug efficacy and providing new strategies for future clinical applications. This study highlights the outlook of nanotechnology in treating neurological disorders caused by heat stroke, offering a novel therapeutic approach with potential clinical applications. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224934","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis 从 TNF-α 预处理 IPFP-MSCs 提取的工程外泌体可提高骨关节炎的产量和疗效
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-11 DOI: 10.1186/s12951-024-02795-9
Jiangyi Wu, Jinhui Wu, Wei Xiang, Yunquan Gong, Daibo Feng, Shunzheng Fang, Yaran Wu, Zheng Liu, Yang Li, Ran Chen, Xiaoqi Zhang, Bingfei Li, Lifeng Chen, Runze Jin, Song Li, Bin Zhang, Tongyi Zhang, Lin Yin, Yizhao Zhou, Shu Huang, Ningning Liu, Hao Xu, Jiqin Lian, Yongqian Wang, Siru Zhou, Zhenhong Ni
The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF−α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF−α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF−α, thereby exerting chondroprotective effects. TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.
骨关节炎(OA)的发病机制包括关节软骨的逐渐退化。从间充质干细胞中提取的外泌体(MSC-EXOs)已被证明可通过减轻软骨破坏来减轻关节病理损伤。优化间充质干细胞外泌体的产量和疗效对促进其临床转化至关重要。间充质干细胞的预处理可增强工程外泌体的治疗潜力,通过可控和可量化的外部刺激,为其应用提供了广阔的前景。本研究旨在通过对间叶干细胞进行促炎预处理来提高外泌体的产生并增强其对 OA 的疗效,从而解决这些问题。研究人员从使用促炎因子 TNF-α 预处理的髌下脂肪垫(IPFP)-间充质干细胞的上清液中分离出外泌体,随后对其产生量进行了量化。在TNF-α预处理前后,通过高通量转录组测序分析、q-PCR和Western印迹分析评估了IPFP-间充质干细胞分泌外泌体的相关途径。此外,还将从TNF-α预处理的IPFP-间充质干细胞中提取的外泌体(IPFP-间充质干细胞-EXOsTNF-α)用于OA小鼠模型的关节内给药,并对关节病理学和步态改变进行了后续评估。通过蛋白质组分析确定了外泌体中参与维持软骨平衡的蛋白质的表达。与未经预处理的间充质干细胞相比,TNF-α的预处理能显著增强IPFP-间充质干细胞的外泌体分泌。其潜在机制是TNF-α预处理激活了IPFP-间充质干细胞的PI3K/AKT信号通路,导致自噬相关蛋白16 like 1(ATG16L1)水平上调,从而促进了外泌体的分泌。IPFP-间充质干细胞-EXOsTNF-α的关节内给药在改善OA小鼠关节病理变化方面表现出卓越的疗效。TNF-α的预处理增强了IPFP-间充质干细胞-EXOsTNF-α中低密度脂蛋白受体相关蛋白1(LRP1)水平的上调,从而发挥了软骨保护作用。TNF-α预处理是优化IPFP-间充质干细胞外泌体治疗OA疗效的一种有效且有前景的方法。
{"title":"Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis","authors":"Jiangyi Wu, Jinhui Wu, Wei Xiang, Yunquan Gong, Daibo Feng, Shunzheng Fang, Yaran Wu, Zheng Liu, Yang Li, Ran Chen, Xiaoqi Zhang, Bingfei Li, Lifeng Chen, Runze Jin, Song Li, Bin Zhang, Tongyi Zhang, Lin Yin, Yizhao Zhou, Shu Huang, Ningning Liu, Hao Xu, Jiqin Lian, Yongqian Wang, Siru Zhou, Zhenhong Ni","doi":"10.1186/s12951-024-02795-9","DOIUrl":"https://doi.org/10.1186/s12951-024-02795-9","url":null,"abstract":"The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF−α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF−α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF−α, thereby exerting chondroprotective effects. TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of size, surface charge, and PEGylated lipids of lipid nanoparticles (LNPs) on intramuscular delivery of mRNA 脂质纳米颗粒(LNPs)的尺寸、表面电荷和 PEG 化脂类对肌肉内输送 mRNA 的作用
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-11 DOI: 10.1186/s12951-024-02812-x
Weiwen Kong, Yuning Wei, Zirong Dong, Wenjuan Liu, Jiaxin Zhao, Yan Huang, Jinlong Yang, Wei Wu, Haisheng He, Jianping Qi
Lipid nanoparticles (LNPs) are currently the most commonly used non-viral gene delivery system. Their physiochemical attributes, encompassing size, charge and surface modifications, significantly affect their behaviors both in vivo and in vitro. Nevertheless, the effects of these properties on the transfection and distribution of LNPs after intramuscular injection remain elusive. In this study, LNPs with varying sizes, lipid-based charges and PEGylated lipids were formulated to study their transfection and in vivo distribution. Luciferase mRNA (mLuc) was entraped in LNPs as a model nucleic acid molecule. Results indicated that smaller-sized LNPs and those with neutral potential presented superior transfection efficiency after intramuscular injection. Surprisingly, the sizes and charges did not exert a notable influence on the in vivo distribution of the LNPs. Furthermore, PEGylated lipids with shorter acyl chains contributed to enhanced transfection efficiency due to their superior cellular uptake and lysosomal escape capabilities. Notably, the mechanisms underlying cellular uptake differed among LNPs containing various types of PEGylated lipids, which was primarily attributed to the length of their acyl chain. Together, these insights underscore the pivotal role of nanoparticle characteristics and PEGylated lipids in the intramuscular route. This study not only fills crucial knowledge gaps but also provides significant directions for the effective delivery of mRNA via LNPs.
脂质纳米颗粒(LNPs)是目前最常用的非病毒基因递送系统。脂质纳米颗粒的物理化学属性,包括尺寸、电荷和表面修饰,对其体内和体外行为都有显著影响。然而,这些属性对 LNPs 肌肉注射后的转染和分布的影响仍然难以捉摸。在本研究中,我们配制了具有不同大小、基于脂质的电荷和 PEG 化脂质的 LNPs,以研究它们的转染和体内分布。以荧光素酶 mRNA(mLuc)为模型核酸分子,将其包裹在 LNPs 中。结果表明,较小尺寸的 LNPs 和具有中性电位的 LNPs 在肌肉注射后具有更高的转染效率。令人惊讶的是,大小和电荷对 LNPs 的体内分布并无明显影响。此外,具有较短酰基链的 PEG 化脂类因其卓越的细胞摄取和溶酶体逃逸能力而提高了转染效率。值得注意的是,含有各种类型 PEG 化脂类的 LNPs 的细胞摄取机制各不相同,这主要归因于其酰基链的长度。这些见解共同强调了纳米粒子特性和 PEG 化脂类在肌内途径中的关键作用。这项研究不仅填补了重要的知识空白,而且为通过 LNPs 有效递送 mRNA 提供了重要方向。
{"title":"Role of size, surface charge, and PEGylated lipids of lipid nanoparticles (LNPs) on intramuscular delivery of mRNA","authors":"Weiwen Kong, Yuning Wei, Zirong Dong, Wenjuan Liu, Jiaxin Zhao, Yan Huang, Jinlong Yang, Wei Wu, Haisheng He, Jianping Qi","doi":"10.1186/s12951-024-02812-x","DOIUrl":"https://doi.org/10.1186/s12951-024-02812-x","url":null,"abstract":"Lipid nanoparticles (LNPs) are currently the most commonly used non-viral gene delivery system. Their physiochemical attributes, encompassing size, charge and surface modifications, significantly affect their behaviors both in vivo and in vitro. Nevertheless, the effects of these properties on the transfection and distribution of LNPs after intramuscular injection remain elusive. In this study, LNPs with varying sizes, lipid-based charges and PEGylated lipids were formulated to study their transfection and in vivo distribution. Luciferase mRNA (mLuc) was entraped in LNPs as a model nucleic acid molecule. Results indicated that smaller-sized LNPs and those with neutral potential presented superior transfection efficiency after intramuscular injection. Surprisingly, the sizes and charges did not exert a notable influence on the in vivo distribution of the LNPs. Furthermore, PEGylated lipids with shorter acyl chains contributed to enhanced transfection efficiency due to their superior cellular uptake and lysosomal escape capabilities. Notably, the mechanisms underlying cellular uptake differed among LNPs containing various types of PEGylated lipids, which was primarily attributed to the length of their acyl chain. Together, these insights underscore the pivotal role of nanoparticle characteristics and PEGylated lipids in the intramuscular route. This study not only fills crucial knowledge gaps but also provides significant directions for the effective delivery of mRNA via LNPs. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ellagic acid-enhanced biocompatibility and bioactivity in multilayer core-shell gold nanoparticles for ameliorating myocardial infarction injury 鞣花酸增强多层核壳金纳米粒子的生物相容性和生物活性,改善心肌梗死损伤
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-11 DOI: 10.1186/s12951-024-02796-8
Xina Yu, Jie Wang, Tiantian Wang, Shanshan Song, Hongna Su, Hui Huang, Pei Luo
Myocardial infarction (MI) is the main contributor to most cardiovascular diseases (CVDs), and the available post-treatment clinical therapeutic options are limited. The development of nanoscale drug delivery systems carrying natural small molecules provides biotherapies that could potentially offer new treatments for reactive oxygen species (ROS)-induced damage in MI. Considering the stability and reduced toxicity of gold-phenolic core-shell nanoparticles, this study aims to develop ellagic acid-functionalized gold nanoparticles (EA-AuNPs) to overcome these limitations. We have successfully synthesized EA-AuNPs with enhanced biocompatibility and bioactivity. These core-shell gold nanoparticles exhibit excellent ROS-scavenging activity and high dispersion. The results from a label-free imaging method on optically transparent zebrafish larvae models and micro-CT imaging in mice indicated that EA-AuNPs enable a favorable excretion-based metabolism without overburdening other organs. EA-AuNPs were subsequently applied in cellular oxidative stress models and MI mouse models. We found that they effectively inhibit the expression of apoptosis-related proteins and the elevation of cardiac enzyme activities, thereby ameliorating oxidative stress injuries in MI mice. Further investigations of oxylipin profiles indicated that EA-AuNPs might alleviate myocardial injury by inhibiting ROS-induced oxylipin level alterations, restoring the perturbed anti-inflammatory oxylipins. These findings collectively emphasized the protective role of EA-AuNPs in myocardial injury, which contributes to the development of innovative gold-phenolic nanoparticles and further advances their potential medical applications.
心肌梗塞(MI)是大多数心血管疾病(CVDs)的主要致病因素,而现有的治疗后临床疗法却很有限。携带天然小分子的纳米级给药系统的开发提供了生物疗法,有可能为活性氧(ROS)诱导的心肌梗死损伤提供新的治疗方法。考虑到金酚类核壳纳米粒子的稳定性和低毒性,本研究旨在开发鞣花酸功能化金纳米粒子(EA-AuNPs),以克服这些局限性。我们成功合成了具有更强生物相容性和生物活性的鞣花酸功能化金纳米粒子(EA-AuNPs)。这些核壳金纳米粒子具有优异的清除 ROS 活性和高分散性。在光学透明斑马鱼幼体模型上进行的无标记成像方法和在小鼠体内进行的显微 CT 成像结果表明,EA-AuNPs 可实现良好的排泄型新陈代谢,而不会对其他器官造成过重负担。随后,我们将 EA-AuNPs 应用于细胞氧化应激模型和 MI 小鼠模型。我们发现它们能有效抑制细胞凋亡相关蛋白的表达和心肌酶活性的升高,从而改善心肌梗死小鼠的氧化应激损伤。对氧化脂蛋白谱的进一步研究表明,EA-AuNPs 可抑制 ROS 诱导的氧化脂蛋白水平变化,恢复被扰乱的抗炎氧化脂蛋白,从而减轻心肌损伤。这些发现共同强调了 EA-AuNPs 在心肌损伤中的保护作用,有助于开发创新的金酚类纳米粒子,并进一步推进其潜在的医学应用。
{"title":"Ellagic acid-enhanced biocompatibility and bioactivity in multilayer core-shell gold nanoparticles for ameliorating myocardial infarction injury","authors":"Xina Yu, Jie Wang, Tiantian Wang, Shanshan Song, Hongna Su, Hui Huang, Pei Luo","doi":"10.1186/s12951-024-02796-8","DOIUrl":"https://doi.org/10.1186/s12951-024-02796-8","url":null,"abstract":"Myocardial infarction (MI) is the main contributor to most cardiovascular diseases (CVDs), and the available post-treatment clinical therapeutic options are limited. The development of nanoscale drug delivery systems carrying natural small molecules provides biotherapies that could potentially offer new treatments for reactive oxygen species (ROS)-induced damage in MI. Considering the stability and reduced toxicity of gold-phenolic core-shell nanoparticles, this study aims to develop ellagic acid-functionalized gold nanoparticles (EA-AuNPs) to overcome these limitations. We have successfully synthesized EA-AuNPs with enhanced biocompatibility and bioactivity. These core-shell gold nanoparticles exhibit excellent ROS-scavenging activity and high dispersion. The results from a label-free imaging method on optically transparent zebrafish larvae models and micro-CT imaging in mice indicated that EA-AuNPs enable a favorable excretion-based metabolism without overburdening other organs. EA-AuNPs were subsequently applied in cellular oxidative stress models and MI mouse models. We found that they effectively inhibit the expression of apoptosis-related proteins and the elevation of cardiac enzyme activities, thereby ameliorating oxidative stress injuries in MI mice. Further investigations of oxylipin profiles indicated that EA-AuNPs might alleviate myocardial injury by inhibiting ROS-induced oxylipin level alterations, restoring the perturbed anti-inflammatory oxylipins. These findings collectively emphasized the protective role of EA-AuNPs in myocardial injury, which contributes to the development of innovative gold-phenolic nanoparticles and further advances their potential medical applications. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224938","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Non-viral approaches in CAR-NK cell engineering: connecting natural killer cell biology and gene delivery CAR-NK 细胞工程中的非病毒方法:连接自然杀伤细胞生物学和基因传递
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-10 DOI: 10.1186/s12951-024-02746-4
Emma M. McErlean, Helen O. McCarthy
Natural Killer (NK) cells are exciting candidates for cancer immunotherapy with potent innate cytotoxicity and distinct advantages over T cells for Chimeric Antigen Receptor (CAR) therapy. Concerns regarding the safety, cost, and scalability of viral vectors has ignited research into non-viral alternatives for gene delivery. This review comprehensively analyses recent advancements and challenges with non-viral genetic modification of NK cells for allogeneic CAR-NK therapies. Non-viral alternatives including electroporation and multifunctional nanoparticles are interrogated with respect to CAR expression and translational responses. Crucially, the link between NK cell biology and design of drug delivery technologies are made, which is essential for development of future non-viral approaches. This review provides valuable insights into the current state of non-viral CAR-NK cell engineering, aimed at realising the full potential of NK cell-based immunotherapies. Non-viral production of “off-the-shelf” CAR-NK cells. 1. NK cells may be purified from donor blood, differentiated from stem cells or produced from immortalised cell lines in the lab. 2. NK-specific CAR design modified from CAR-T designs to include NK transmembrane domains (NKG2D, NKp44), co-stimulatory receptors (e.g., DAP10, 2B4) and NK cell receptors (NKG2D). 3. Non-viral genetic modification of NK cells can include delivery of CAR construct via DNA or mRNA, and knock-in/out of specific genes using gene editing tools (e.g., CRISPR Cas9, transposons). This requires a gene delivery method which may include electroporation, lipid and multifunctional nanoparticles and cell penetrating peptides. The resultant CAR-NK cells are then expanded in vitro and may be delivered as an "off-the-shelf" product to treat multiple patients.
自然杀伤(NK)细胞是癌症免疫疗法中令人兴奋的候选细胞,具有强大的先天细胞毒性,在嵌合抗原受体(CAR)疗法中与 T 细胞相比具有明显优势。人们对病毒载体的安全性、成本和可扩展性的担忧点燃了对基因递送非病毒替代品的研究。本综述全面分析了用于异体 CAR-NK 疗法的 NK 细胞非病毒基因改造的最新进展和挑战。非病毒替代方法包括电穿孔和多功能纳米粒子,它们在 CAR 表达和转化反应方面均有研究。最重要的是,该综述将 NK 细胞生物学与给药技术设计联系起来,这对未来非病毒疗法的开发至关重要。这篇综述为非病毒 CAR-NK 细胞工程的现状提供了宝贵的见解,旨在充分发挥基于 NK 细胞的免疫疗法的潜力。非病毒生产 "现成的 "CAR-NK 细胞。1.NK 细胞可以从供体血液中纯化、从干细胞中分化或在实验室中从永生细胞系中产生。2.根据 CAR-T 设计修改 NK 特异性 CAR 设计,以包括 NK 跨膜结构域(NKG2D、NKp44)、共刺激受体(如 DAP10、2B4)和 NK 细胞受体(NKG2D)。3.3. NK 细胞的非病毒基因改造可包括通过 DNA 或 mRNA 运送 CAR 构建物,以及使用基因编辑工具(如 CRISPR Cas9、转座子)敲入/敲出特定基因。这需要一种基因递送方法,其中可能包括电穿孔、脂质和多功能纳米颗粒以及细胞穿透肽。由此产生的 CAR-NK 细胞随后在体外扩增,并可作为 "现成 "产品用于治疗多名患者。
{"title":"Non-viral approaches in CAR-NK cell engineering: connecting natural killer cell biology and gene delivery","authors":"Emma M. McErlean, Helen O. McCarthy","doi":"10.1186/s12951-024-02746-4","DOIUrl":"https://doi.org/10.1186/s12951-024-02746-4","url":null,"abstract":"Natural Killer (NK) cells are exciting candidates for cancer immunotherapy with potent innate cytotoxicity and distinct advantages over T cells for Chimeric Antigen Receptor (CAR) therapy. Concerns regarding the safety, cost, and scalability of viral vectors has ignited research into non-viral alternatives for gene delivery. This review comprehensively analyses recent advancements and challenges with non-viral genetic modification of NK cells for allogeneic CAR-NK therapies. Non-viral alternatives including electroporation and multifunctional nanoparticles are interrogated with respect to CAR expression and translational responses. Crucially, the link between NK cell biology and design of drug delivery technologies are made, which is essential for development of future non-viral approaches. This review provides valuable insights into the current state of non-viral CAR-NK cell engineering, aimed at realising the full potential of NK cell-based immunotherapies. Non-viral production of “off-the-shelf” CAR-NK cells. 1. NK cells may be purified from donor blood, differentiated from stem cells or produced from immortalised cell lines in the lab. 2. NK-specific CAR design modified from CAR-T designs to include NK transmembrane domains (NKG2D, NKp44), co-stimulatory receptors (e.g., DAP10, 2B4) and NK cell receptors (NKG2D). 3. Non-viral genetic modification of NK cells can include delivery of CAR construct via DNA or mRNA, and knock-in/out of specific genes using gene editing tools (e.g., CRISPR Cas9, transposons). This requires a gene delivery method which may include electroporation, lipid and multifunctional nanoparticles and cell penetrating peptides. The resultant CAR-NK cells are then expanded in vitro and may be delivered as an \"off-the-shelf\" product to treat multiple patients. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224941","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rationally designed catalytic nanoplatform for enhanced chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment 合理设计催化纳米平台,通过部署内源性和外源性铜并重塑肿瘤微环境来增强化学免疫疗法
IF 10.2 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-09 DOI: 10.1186/s12951-024-02696-x
Daxi Sun, Liting Yu, Gang Wang, Yuxue Xu, Peng Wang, Ningning Wang, Zhengyan Wu, Guilong Zhang, Jia Zhang, Yunjiao Zhang, Geng Tian, Pengfei Wei
Chemodynamic therapy represents a novel tumor therapeutic modality via triggering catalytic reactions in tumors to yield highly toxic reactive oxygen species (ROS). Nevertheless, low efficiency catalytic ability, potential systemic toxicity and inefficient tumor targeting, have hindered the efficacy of chemodynamic therapy. Herein, a rationally designed catalytic nanoplatform, composed of folate acid conjugated liposomes loaded with copper peroxide (CP) and chloroquine (CQ; a clinical drug) (denoted as CC@LPF), could power maximal tumor cytotoxicity, mechanistically via maneuvering endogenous and exogenous copper for a highly efficient catalytic reaction. Despite a massive autophagosome accumulation elicited by CP-powered autophagic initiation and CQ-induced autolysosomal blockage, the robust ROS, but not aberrant autophagy, underlies the synergistic tumor inhibition. Otherwise, this combined mode also elicits an early onset, above all, long-term high-level existence of immunogenic cell death markers, associated with ROS and aberrant autophagy -triggered endoplasmic reticulum stress. Besides, CC@LPF, with tumor targeting capability and selective tumor cytotoxicity, could elicit intratumor dendritic cells (mainly attributed to CQ) and tumor infiltrating CD8+ T cells, upon combining with PD-L1 therapeutic antibody, further induce significant anti-tumor effect. Collectively, the rationally designed nanoplatform, CC@LPF, could enhance tumor chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment.
化学动力疗法是一种新型的肿瘤治疗方法,它通过触发肿瘤内的催化反应产生高毒性活性氧(ROS)。然而,低效催化能力、潜在的全身毒性和低效的肿瘤靶向性阻碍了化学动力疗法的疗效。在这里,一种合理设计的催化纳米平台由叶酸共轭脂质体组成,内含过氧化铜(CP)和氯喹(CQ,一种临床药物)(称为 CC@LPF),可通过操纵内源性和外源性铜进行高效催化反应,从机理上发挥最大的肿瘤细胞毒性。尽管 CP 促成的自噬启动和 CQ 诱导的自溶酶体阻断引起了大量的自噬体积累,但强大的 ROS 而非异常的自噬是协同抑制肿瘤的基础。否则,这种联合模式也会导致免疫原性细胞死亡标志物的早期出现,尤其是长期高水平的存在,这与 ROS 和异常自噬触发的内质网应激有关。此外,CC@LPF具有肿瘤靶向能力和选择性肿瘤细胞毒性,可诱导肿瘤内树突状细胞(主要是CQ)和肿瘤浸润的CD8+T细胞,与PD-L1治疗抗体结合后可进一步诱导显著的抗肿瘤效果。总之,合理设计的纳米平台CC@LPF可通过调配内源性和外源性铜并重塑肿瘤微环境来增强肿瘤化疗免疫疗法。
{"title":"Rationally designed catalytic nanoplatform for enhanced chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment","authors":"Daxi Sun, Liting Yu, Gang Wang, Yuxue Xu, Peng Wang, Ningning Wang, Zhengyan Wu, Guilong Zhang, Jia Zhang, Yunjiao Zhang, Geng Tian, Pengfei Wei","doi":"10.1186/s12951-024-02696-x","DOIUrl":"https://doi.org/10.1186/s12951-024-02696-x","url":null,"abstract":"Chemodynamic therapy represents a novel tumor therapeutic modality via triggering catalytic reactions in tumors to yield highly toxic reactive oxygen species (ROS). Nevertheless, low efficiency catalytic ability, potential systemic toxicity and inefficient tumor targeting, have hindered the efficacy of chemodynamic therapy. Herein, a rationally designed catalytic nanoplatform, composed of folate acid conjugated liposomes loaded with copper peroxide (CP) and chloroquine (CQ; a clinical drug) (denoted as CC@LPF), could power maximal tumor cytotoxicity, mechanistically via maneuvering endogenous and exogenous copper for a highly efficient catalytic reaction. Despite a massive autophagosome accumulation elicited by CP-powered autophagic initiation and CQ-induced autolysosomal blockage, the robust ROS, but not aberrant autophagy, underlies the synergistic tumor inhibition. Otherwise, this combined mode also elicits an early onset, above all, long-term high-level existence of immunogenic cell death markers, associated with ROS and aberrant autophagy -triggered endoplasmic reticulum stress. Besides, CC@LPF, with tumor targeting capability and selective tumor cytotoxicity, could elicit intratumor dendritic cells (mainly attributed to CQ) and tumor infiltrating CD8+ T cells, upon combining with PD-L1 therapeutic antibody, further induce significant anti-tumor effect. Collectively, the rationally designed nanoplatform, CC@LPF, could enhance tumor chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.2,"publicationDate":"2024-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142224952","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Apoptosis and cuproptosis Co-activated Copper-based metal-organic frameworks for cancer therapy. 用于癌症治疗的铜基金属有机框架的凋亡和杯突变共激活。
IF 10.6 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-06 DOI: 10.1186/s12951-024-02828-3
Kun Li, Leilei Wu, Han Wang, Zi Fu, Jiani Gao, Xiucheng Liu, Yongfei Fan, Xichun Qin, Dalong Ni, Jing Wang, Dong Xie

Lung cancer, predominantly non-small cell lung cancer (NSCLC), remains a significant global health challenge, with limited therapeutic options for patients with KRAS-mutated tumors. Herein, a copper-based metal-organic framework (Cu-MOF) was applied as a novel cuproptosis-mediated nanoplatform for lung cancer therapy. Cu-MOF would disassemble and liberate copper ions under the acidic microenvironment of lysosomes of cancer cells, initiating a cascade of cellular events. The released copper ions catalyzes the Fenton reaction, generating hydroxyl radicals that induce oxidative damage, leading to cytoskeletal disruption and activation of caspase-3, ultimately triggering apoptosis. Simultaneously, with the mediation of the key regulatory factor FDX1, we found that the copper ions binding to the mitochondrial protein DLAT could result in the loss of iron-sulfur cluster proteins and aggregation of lipoylated proteins, which culminated in proteotoxic stress-induced cuproptosis. The pronounced anti-tumor effects of Cu-MOF with apoptosis and cuproptosis were confirmed both in vitro and in vivo experiments. Such dual induction of apoptosis and cuproptosis by Cu-MOF presents a promising therapeutic strategy for NSCLC, particularly for KRAS-mutated tumors, and expands potential applications of Cu-based nanomateirals for other cancers.

肺癌,主要是非小细胞肺癌(NSCLC),仍然是全球健康面临的重大挑战,而 KRAS 基因突变肿瘤患者的治疗选择有限。在本文中,铜基金属有机框架(Cu-MOF)被用作一种新型的杯突症介导的肺癌治疗纳米平台。在癌细胞溶酶体的酸性微环境下,Cu-MOF 会解体并释放出铜离子,从而引发一系列细胞事件。释放出的铜离子催化芬顿反应,产生羟自由基,诱发氧化损伤,导致细胞骨架破坏和激活 Caspase-3,最终引发细胞凋亡。同时,在关键调控因子 FDX1 的介导下,我们发现铜离子与线粒体蛋白 DLAT 的结合可导致铁硫簇蛋白的缺失和脂酰化蛋白的聚集,最终导致蛋白毒性应激诱导的杯突症。体外和体内实验都证实了 Cu-MOF 在凋亡和杯突变方面的显著抗肿瘤作用。Cu-MOF 的这种凋亡和杯突变双重诱导作用为治疗 NSCLC(尤其是 KRAS 突变肿瘤)提供了一种前景广阔的治疗策略,并拓展了铜基纳米药物在其他癌症中的潜在应用。
{"title":"Apoptosis and cuproptosis Co-activated Copper-based metal-organic frameworks for cancer therapy.","authors":"Kun Li, Leilei Wu, Han Wang, Zi Fu, Jiani Gao, Xiucheng Liu, Yongfei Fan, Xichun Qin, Dalong Ni, Jing Wang, Dong Xie","doi":"10.1186/s12951-024-02828-3","DOIUrl":"10.1186/s12951-024-02828-3","url":null,"abstract":"<p><p>Lung cancer, predominantly non-small cell lung cancer (NSCLC), remains a significant global health challenge, with limited therapeutic options for patients with KRAS-mutated tumors. Herein, a copper-based metal-organic framework (Cu-MOF) was applied as a novel cuproptosis-mediated nanoplatform for lung cancer therapy. Cu-MOF would disassemble and liberate copper ions under the acidic microenvironment of lysosomes of cancer cells, initiating a cascade of cellular events. The released copper ions catalyzes the Fenton reaction, generating hydroxyl radicals that induce oxidative damage, leading to cytoskeletal disruption and activation of caspase-3, ultimately triggering apoptosis. Simultaneously, with the mediation of the key regulatory factor FDX1, we found that the copper ions binding to the mitochondrial protein DLAT could result in the loss of iron-sulfur cluster proteins and aggregation of lipoylated proteins, which culminated in proteotoxic stress-induced cuproptosis. The pronounced anti-tumor effects of Cu-MOF with apoptosis and cuproptosis were confirmed both in vitro and in vivo experiments. Such dual induction of apoptosis and cuproptosis by Cu-MOF presents a promising therapeutic strategy for NSCLC, particularly for KRAS-mutated tumors, and expands potential applications of Cu-based nanomateirals for other cancers.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.6,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11378619/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142140334","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Nanobiotechnology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1