首页 > 最新文献

Biomaterials最新文献

英文 中文
Transplantation of peripheral nerve tissueoid based on a decellularized optic nerve scaffold to restore rat hindlimb sensory and movement functions 基于脱细胞视神经支架的外周神经组织样体移植用于恢复大鼠后肢的感觉和运动功能。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-06 DOI: 10.1016/j.biomaterials.2024.122949
Zhao-Wei Zhu , Ge Li , Guang-Geng Wu , Yu-Jing Zhang , Yu-Rong Bai , Bi-Qin Lai , Ying Ding , Xiang Zeng , Yuan-Huan Ma , Shu Liu , Rui Wang , Jing-Hua Liang , Yang-Bin Xu , Bo He , Yuan-Shan Zeng
Peripheral nerve injury (PNI) involving the loss of sensory and movement functions is challenging to repair. Although the gold standard of PNI repair is still the use of autologous nerve grafts, the destruction of the donor side is inevitable. In the present study, peripheral nerve tissueoids (PNTs) composed of a Schwann cell (SC)-based neurotrophin-3 (NT-3) delivery system and a decellularized optic nerve (DON) with naturally oriented channels were engineered to investigate the mechanism of PNTs in nerve regeneration. Proteomic analysis and mRNA sequencing revealed that PNTs have the advantage of promoting nerve regeneration by the three mechanisms of chemotaxis, adhesion and intrinsic mobilisation. The results demonstrated that a local NT-3-enriched pool was constructed by laminin γ3 (LAMC3) in PNTs, creating a niche for the colonization of TrkC-positive SCs, attraction of axons to the defect/graft area, and remyelination. In addition, LAMC3 in PNTs can rapidly promote axon adhesion through integrin aVβ6 and can precisely guide long projecting axons to target tissues. Furthermore, the interactions among the NT-3/TrkC, LAMC3/integrin aVβ6 and the scaffold synergistically activate the PI3K-AKT signalling pathway in damaged neurons, further stimulating the intrinsic regenerative drive within the neurons to ultimately achieve the rapid reinnervation and the improvement of sensory and movement functions in the hindlimb.
周围神经损伤(PNI)涉及感觉和运动功能的丧失,修复难度很大。虽然自体神经移植仍是修复周围神经损伤的金标准,但供体侧的破坏是不可避免的。本研究设计了由基于许旺细胞(SC)的神经营养素-3(NT-3)递送系统和具有自然定向通道的脱细胞视神经(DON)组成的外周神经组织实体(PNTs),以研究 PNTs 在神经再生中的作用机制。蛋白质组分析和 mRNA 测序显示,PNTs 具有通过趋化、粘附和内在调动三种机制促进神经再生的优势。结果表明,层粘蛋白γ3(LAMC3)在PNTs中构建了一个富含NT-3的局部池,为TrkC阳性SCs的定植、轴突被吸引到缺损/移植物区域以及再髓鞘化创造了有利条件。此外,PNTs 中的 LAMC3 还能通过整合素 aVβ6 快速促进轴突粘附,并能精确引导长轴突到达靶组织。此外,NT-3/TrkC、LAMC3/整合素 aVβ6 和支架之间的相互作用可协同激活受损神经元中的 PI3K-AKT 信号通路,进一步激发神经元的内在再生动力,最终实现快速神经支配,改善后肢的感觉和运动功能。
{"title":"Transplantation of peripheral nerve tissueoid based on a decellularized optic nerve scaffold to restore rat hindlimb sensory and movement functions","authors":"Zhao-Wei Zhu ,&nbsp;Ge Li ,&nbsp;Guang-Geng Wu ,&nbsp;Yu-Jing Zhang ,&nbsp;Yu-Rong Bai ,&nbsp;Bi-Qin Lai ,&nbsp;Ying Ding ,&nbsp;Xiang Zeng ,&nbsp;Yuan-Huan Ma ,&nbsp;Shu Liu ,&nbsp;Rui Wang ,&nbsp;Jing-Hua Liang ,&nbsp;Yang-Bin Xu ,&nbsp;Bo He ,&nbsp;Yuan-Shan Zeng","doi":"10.1016/j.biomaterials.2024.122949","DOIUrl":"10.1016/j.biomaterials.2024.122949","url":null,"abstract":"<div><div>Peripheral nerve injury (PNI) involving the loss of sensory and movement functions is challenging to repair. Although the gold standard of PNI repair is still the use of autologous nerve grafts, the destruction of the donor side is inevitable. In the present study, peripheral nerve tissueoids (PNTs) composed of a Schwann cell (SC)-based neurotrophin-3 (NT-3) delivery system and a decellularized optic nerve (DON) with naturally oriented channels were engineered to investigate the mechanism of PNTs in nerve regeneration. Proteomic analysis and mRNA sequencing revealed that PNTs have the advantage of promoting nerve regeneration by the three mechanisms of chemotaxis, adhesion and intrinsic mobilisation. The results demonstrated that a local NT-3-enriched pool was constructed by laminin γ3 (LAMC3) in PNTs, creating a niche for the colonization of TrkC-positive SCs, attraction of axons to the defect/graft area, and remyelination. In addition, LAMC3 in PNTs can rapidly promote axon adhesion through integrin aVβ6 and can precisely guide long projecting axons to target tissues. Furthermore, the interactions among the NT-3/TrkC, LAMC3/integrin aVβ6 and the scaffold synergistically activate the PI3K-AKT signalling pathway in damaged neurons, further stimulating the intrinsic regenerative drive within the neurons to ultimately achieve the rapid reinnervation and the improvement of sensory and movement functions in the hindlimb.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122949"},"PeriodicalIF":12.8,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142613394","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A 3D-printed microdevice encapsulates vascularized islets composed of iPSC-derived β-like cells and microvascular fragments for type 1 diabetes treatment 一种三维打印微型装置封装了由源自iPSC的β样细胞和微血管片段组成的血管化小胰岛,用于治疗1型糖尿病。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-06 DOI: 10.1016/j.biomaterials.2024.122947
Shuang Chen, Wenshuang Wang, Lanlin Shen, Haofan Liu, Jing Luo, Yushuang Ren, Susu Cui, Yixin Ye, Gang Shi, Fuyi Cheng, Xiaolan Su, Lei Dai, Maling Gou, Hongxin Deng
Transplantation of insulin-secreting cells provides a promising method for re-establishing the autonomous blood glucose control ability of type 1 diabetes (T1D) patients, but the low survival of the transplanted cells hinder the therapeutic efficacy. In this study, we 3D-printed an encapsulation system containing β-like cells and microvascular fragments (MVF), to create a retrivable microdevice with vascularized islets in vivo for T1D therapy. The functional β-like cells were differentiated from the urine epithelial cell-derived induced pluripotent stem cells (UiPSCs). Single-cell RNA sequencing provided an integrative study and macroscopic developmental analyses of the entire process of differentiation, which revealed the developmental trajectory of differentiation in vitro follows the developmental pattern of embryonic pancreas in vivo. The MVF were isolated from the epididymal fat pad. The microdevice with a groove structure were rapidly fabricated by the digital light processing (DLP)-3D printing technology. The β-like cells and MVF were uniformly distributed in the device. After subcutaneous transplantation into C57BL/6 mice, the microdevice have less collagen accumulation and low immune cell infiltration. Moreover, the microdevice encapsulated vascularized islets reduced hyperglycemia in 33 % of the treated mice for up to 100 days without immunosuppressants, and the humanized C-peptide was also detected in the serum of the mice. In summary, we described the microdevice-protected vascularized islets for long-term treatment of T1D, with high safety and potential clinical transformative value, and may therefore provide a translatable solution to advance the research progress of β cell replacement therapy for T1D.
胰岛素分泌细胞的移植为重建1型糖尿病(T1D)患者的自主血糖控制能力提供了一种可行的方法,但移植细胞的低存活率阻碍了治疗效果。在这项研究中,我们用三维打印技术制造了一种含有β样细胞和微血管片段(MVF)的封装系统,从而在体内制造出一种带有血管化小胰岛的可重复使用的微型装置,用于治疗T1D。功能性β样细胞由尿液上皮细胞衍生的诱导多能干细胞(UiPSCs)分化而来。单细胞 RNA 测序对整个分化过程进行了综合研究和宏观发育分析,发现体外分化的发育轨迹与体内胚胎胰腺的发育模式一致。MVF是从附睾脂肪垫中分离出来的。利用数字光处理(DLP)-三维打印技术快速制备了具有凹槽结构的微器件。β样细胞和MVF均匀地分布在装置中。将微装置移植到 C57BL/6 小鼠皮下后,胶原堆积较少,免疫细胞浸润较低。此外,在不使用免疫抑制剂的情况下,封装了血管化胰岛的微装置在长达 100 天的治疗中减少了 33% 的小鼠的高血糖症状,而且在小鼠血清中也检测到了人源化 C 肽。总之,我们描述的微装置保护血管化小胰岛用于T1D的长期治疗,具有高度的安全性和潜在的临床转化价值,因此可能为推动T1D的β细胞替代疗法的研究进展提供一种可转化的解决方案。
{"title":"A 3D-printed microdevice encapsulates vascularized islets composed of iPSC-derived β-like cells and microvascular fragments for type 1 diabetes treatment","authors":"Shuang Chen,&nbsp;Wenshuang Wang,&nbsp;Lanlin Shen,&nbsp;Haofan Liu,&nbsp;Jing Luo,&nbsp;Yushuang Ren,&nbsp;Susu Cui,&nbsp;Yixin Ye,&nbsp;Gang Shi,&nbsp;Fuyi Cheng,&nbsp;Xiaolan Su,&nbsp;Lei Dai,&nbsp;Maling Gou,&nbsp;Hongxin Deng","doi":"10.1016/j.biomaterials.2024.122947","DOIUrl":"10.1016/j.biomaterials.2024.122947","url":null,"abstract":"<div><div>Transplantation of insulin-secreting cells provides a promising method for re-establishing the autonomous blood glucose control ability of type 1 diabetes (T1D) patients, but the low survival of the transplanted cells hinder the therapeutic efficacy. In this study, we 3D-printed an encapsulation system containing β-like cells and microvascular fragments (MVF), to create a retrivable microdevice with vascularized islets in vivo for T1D therapy. The functional β-like cells were differentiated from the urine epithelial cell-derived induced pluripotent stem cells (UiPSCs). Single-cell RNA sequencing provided an integrative study and macroscopic developmental analyses of the entire process of differentiation, which revealed the developmental trajectory of differentiation in vitro follows the developmental pattern of embryonic pancreas in vivo. The MVF were isolated from the epididymal fat pad. The microdevice with a groove structure were rapidly fabricated by the digital light processing (DLP)-3D printing technology. The β-like cells and MVF were uniformly distributed in the device. After subcutaneous transplantation into C57BL/6 mice, the microdevice have less collagen accumulation and low immune cell infiltration. Moreover, the microdevice encapsulated vascularized islets reduced hyperglycemia in 33 % of the treated mice for up to 100 days without immunosuppressants, and the humanized C-peptide was also detected in the serum of the mice. In summary, we described the microdevice-protected vascularized islets for long-term treatment of T1D, with high safety and potential clinical transformative value, and may therefore provide a translatable solution to advance the research progress of β cell replacement therapy for T1D.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122947"},"PeriodicalIF":12.8,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142637983","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
UC-MSCs based on biomimetic microniche exert excellent regulatory effects on acute brain inflammation through advantageous properties 基于生物仿生微粒的 UC 间充质干细胞通过其优势特性对急性脑部炎症产生了良好的调节作用。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-05 DOI: 10.1016/j.biomaterials.2024.122945
Bichun Zhao , Chao Wang , Manqiang Sun , Xiaocao Ma , Quan Zeng , Jiafei Xi , Junnian Zhou , Xuetao Pei , Yali Jia , Wen Yue
Neuroinflammation triggered by activated microglia leads to neuronal damage and, to a certain extent, neurodegeneration. Human umbilical cord mesenchymal stem cells (UC-MSCs) have good immunomodulatory and neuroprotective effects as well as therapeutic potential for neuroinflammation-related diseases. However, the complex microenvironment created by neuroinflammation poses a challenge to transplanted UC-MSCs. The emerging biomimetic microniche (BN)-based culture technology provides new opportunities to optimize the preparation of UC-MSCs; but the fundamental changes in the characteristics of UC-MSCs based on BN remain unclear, and more reliable preclinical data are needed to support their ability to regulate inflammation. Here, we systematically studied the cellular properties and inflammation regulatory capacity of UC-MSCs in conventional static planar culture (SP-UCMSCs) and suspension culture based on BN (BN-UCMSCs). In vitro, compared with SP-UCMSCs, BN-UCMSCs not only maintained the fundamental characteristics of MSCs, but also significantly enhanced cell proliferation, adhesion, and migration capabilities, etc; notably, the paracrine function and anti-inflammatory capacity of BN-UCMSCs were also enhanced. We further established a murine model of acute brain inflammation and demonstrated that the expression level of pro-inflammatory cytokines in hippocampal and cortical tissues of the BN-UCMSCs group was significantly decreased compared with that in the SP-UCMSCs group. Subsequent transcriptomic analysis of hippocampal and cortical tissues revealed that BN-UCMSCs had the advantage of significantly reducing the expression of pro-inflammatory cytokines through the TLR4-Myd88-NF-κB axis, which was further validated at the gene and protein levels. Taken together, these data strongly indicated that BN-UCMSCs exerts excellent regulatory effects on acute brain inflammation through advantageous properties.
活化的小胶质细胞引发的神经炎症会导致神经元损伤,并在一定程度上导致神经变性。人脐带间充质干细胞(UC-MSCs)具有良好的免疫调节和神经保护作用,并具有治疗神经炎症相关疾病的潜力。然而,神经炎症造成的复杂微环境对移植的 UC-MSCs 构成了挑战。新兴的基于仿生微米(BN)的培养技术为优化 UC 间充质干细胞的制备提供了新的机会;但基于 BN 的 UC 间充质干细胞特性的根本变化仍不清楚,需要更可靠的临床前数据来支持其调节炎症的能力。在这里,我们系统研究了传统静态平面培养(SP-UCMSCs)和基于 BN 的悬浮培养(BN-UCMSCs)中 UC-MSCs 的细胞特性和炎症调节能力。在体外,与 SP-UCMSCs 相比,BN-UCMSCs 不仅保持了间充质干细胞的基本特性,还显著增强了细胞增殖、粘附和迁移等能力;特别是,BN-UCMSCs 的旁分泌功能和抗炎能力也得到了增强。我们进一步建立了小鼠急性脑部炎症模型,结果表明,与 SP-UCMSCs 组相比,BN-UCMSCs 组海马和皮质组织中促炎细胞因子的表达水平明显降低。随后对海马和大脑皮层组织进行的转录组学分析表明,BN-UCMSCs 具有通过 TLR4-Myd88-NF-κB 轴显著减少促炎细胞因子表达的优势,这一点在基因和蛋白质水平上得到了进一步验证。总之,这些数据有力地表明,BN-UCMSCs 通过其优势特性对急性脑部炎症产生了良好的调节作用。
{"title":"UC-MSCs based on biomimetic microniche exert excellent regulatory effects on acute brain inflammation through advantageous properties","authors":"Bichun Zhao ,&nbsp;Chao Wang ,&nbsp;Manqiang Sun ,&nbsp;Xiaocao Ma ,&nbsp;Quan Zeng ,&nbsp;Jiafei Xi ,&nbsp;Junnian Zhou ,&nbsp;Xuetao Pei ,&nbsp;Yali Jia ,&nbsp;Wen Yue","doi":"10.1016/j.biomaterials.2024.122945","DOIUrl":"10.1016/j.biomaterials.2024.122945","url":null,"abstract":"<div><div>Neuroinflammation triggered by activated microglia leads to neuronal damage and, to a certain extent, neurodegeneration. Human umbilical cord mesenchymal stem cells (UC-MSCs) have good immunomodulatory and neuroprotective effects as well as therapeutic potential for neuroinflammation-related diseases. However, the complex microenvironment created by neuroinflammation poses a challenge to transplanted UC-MSCs. The emerging biomimetic microniche (BN)-based culture technology provides new opportunities to optimize the preparation of UC-MSCs; but the fundamental changes in the characteristics of UC-MSCs based on BN remain unclear, and more reliable preclinical data are needed to support their ability to regulate inflammation. Here, we systematically studied the cellular properties and inflammation regulatory capacity of UC-MSCs in conventional static planar culture (SP-UCMSCs) and suspension culture based on BN (BN-UCMSCs). <em>In vitro</em>, compared with SP-UCMSCs, BN-UCMSCs not only maintained the fundamental characteristics of MSCs, but also significantly enhanced cell proliferation, adhesion, and migration capabilities, etc; notably, the paracrine function and anti-inflammatory capacity of BN-UCMSCs were also enhanced. We further established a murine model of acute brain inflammation and demonstrated that the expression level of pro-inflammatory cytokines in hippocampal and cortical tissues of the BN-UCMSCs group was significantly decreased compared with that in the SP-UCMSCs group. Subsequent transcriptomic analysis of hippocampal and cortical tissues revealed that BN-UCMSCs had the advantage of significantly reducing the expression of pro-inflammatory cytokines through the TLR4-Myd88-NF-κB axis, which was further validated at the gene and protein levels. Taken together, these data strongly indicated that BN-UCMSCs exerts excellent regulatory effects on acute brain inflammation through advantageous properties.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122945"},"PeriodicalIF":12.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142613395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rapid and efficient immune response induced by a designed modular cholera toxin B subunit (CTB)-based self-assembling nanoparticle 基于设计的霍乱毒素 B 亚基(CTB)模块化自组装纳米粒子诱导快速高效的免疫反应。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-05 DOI: 10.1016/j.biomaterials.2024.122946
Chao Pan , Shujuan Yu , Caixia Li , Juntao Li , Peng Sun , Yan Guo , Ting Li , Dongshu Wang , Kangfeng Wang , Yufei Lyu , Xiankai Liu , Xiang Li , Jun Wu , Li Zhu , Hengliang Wang
Modular self-assembling nanoparticle vaccines, represent a cutting-edge approach in immunology with the potential to revolutionize vaccine design and efficacy. Although many innovative efficient modular self-assembling nanoparticles have been designed for vaccination, the immune activation characteristics underlying such strong protection remain poorly understood, limiting the further expansion of such nanocarrier. Here, we prepared a novel modular nanovaccine, which self-assembled via a pentamer cholera toxin B subunit (CTB) domain and an unnatural trimer domain, presenting S. Paratyphi A O-polysaccharide antigen, and investigated its rapid immune activation mechanism. The nanovaccine efficiently targets draining lymph nodes and antigen-presenting cells, facilitating co-localization with Golgi and endoplasmic reticulum. In addition, dendritic cells, macrophages, B cells, and neutrophils potentially participate in antigen presentation, unveiling a dynamic change of the vaccines in lymph nodes. Single-cell RNA sequencing at early stage and iN vivo/iN vitro experiments reveal its potent humoral immune response capabilities and protection effects. This nanoparticle outperforms traditional CTB carriers in eliciting robust prophylactic effects in various infection models. This work not only provides a promising and efficient candidate vaccine, but also promotes the design and application of the new type of self-assembled nanoparticle, offering a safe and promising vaccination strategy for infection diseases.
模块化自组装纳米粒子疫苗是免疫学的前沿方法,有可能彻底改变疫苗的设计和功效。尽管已经设计出了许多创新的高效模块化自组装纳米颗粒用于疫苗接种,但人们对这种强保护性所依赖的免疫激活特性仍然知之甚少,从而限制了这种纳米载体的进一步推广。在此,我们制备了一种新型模块化纳米疫苗,它通过五聚体霍乱毒素 B 亚基(CTB)结构域和非天然三聚体结构域自组装,呈现副伤寒甲型流感杆菌 O 型多糖抗原,并研究了其快速免疫激活机制。该纳米疫苗可有效靶向引流淋巴结和抗原递呈细胞,促进与高尔基体和内质网的共定位。此外,树突状细胞、巨噬细胞、B 细胞和中性粒细胞也可能参与抗原呈递,从而揭示了疫苗在淋巴结中的动态变化。早期的单细胞 RNA 测序和体内/体外实验揭示了其强大的体液免疫反应能力和保护效果。在各种感染模型中,这种纳米粒子在激发强大的预防效果方面优于传统的 CTB 载体。这项工作不仅提供了一种有前景的高效候选疫苗,而且促进了新型自组装纳米粒子的设计和应用,为感染性疾病提供了一种安全、有前景的疫苗接种策略。
{"title":"Rapid and efficient immune response induced by a designed modular cholera toxin B subunit (CTB)-based self-assembling nanoparticle","authors":"Chao Pan ,&nbsp;Shujuan Yu ,&nbsp;Caixia Li ,&nbsp;Juntao Li ,&nbsp;Peng Sun ,&nbsp;Yan Guo ,&nbsp;Ting Li ,&nbsp;Dongshu Wang ,&nbsp;Kangfeng Wang ,&nbsp;Yufei Lyu ,&nbsp;Xiankai Liu ,&nbsp;Xiang Li ,&nbsp;Jun Wu ,&nbsp;Li Zhu ,&nbsp;Hengliang Wang","doi":"10.1016/j.biomaterials.2024.122946","DOIUrl":"10.1016/j.biomaterials.2024.122946","url":null,"abstract":"<div><div>Modular self-assembling nanoparticle vaccines, represent a cutting-edge approach in immunology with the potential to revolutionize vaccine design and efficacy. Although many innovative efficient modular self-assembling nanoparticles have been designed for vaccination, the immune activation characteristics underlying such strong protection remain poorly understood, limiting the further expansion of such nanocarrier. Here, we prepared a novel modular nanovaccine, which self-assembled via a pentamer cholera toxin B subunit (CTB) domain and an unnatural trimer domain, presenting <em>S.</em> Paratyphi A O-polysaccharide antigen, and investigated its rapid immune activation mechanism. The nanovaccine efficiently targets draining lymph nodes and antigen-presenting cells, facilitating co-localization with Golgi and endoplasmic reticulum. In addition, dendritic cells, macrophages, B cells, and neutrophils potentially participate in antigen presentation, unveiling a dynamic change of the vaccines in lymph nodes. Single-cell RNA sequencing at early stage and <em>i</em>N <em>vivo</em>/<em>i</em>N <em>vitro</em> experiments reveal its potent humoral immune response capabilities and protection effects. This nanoparticle outperforms traditional CTB carriers in eliciting robust prophylactic effects in various infection models. This work not only provides a promising and efficient candidate vaccine, but also promotes the design and application of the new type of self-assembled nanoparticle, offering a safe and promising vaccination strategy for infection diseases.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122946"},"PeriodicalIF":12.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineering M2 macrophage-derived exosomes modulate activated T cell cuproptosis to promote immune tolerance in rheumatoid arthritis 工程M2巨噬细胞衍生的外泌体调节活化T细胞杯突,促进类风湿性关节炎的免疫耐受。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-02 DOI: 10.1016/j.biomaterials.2024.122943
Guoquan Wu , Tianyu Su , Peng Zhou , Rongze Tang , Xu Zhu , Jin Wang , Minghao Chao , Liying Fan , Hanrong Yan , Peng Ye , Dehong Yu , Fenglei Gao , Hongliang Chen
Nanomedicines for immune modulation have made advancements in the treatment of rheumatoid arthritis (RA). However, due to aberrations in patients' immune systems, inducing antigen-specific immune tolerance while halting disease progression remains a significant challenge. Here, we develop a highly targeted multifunctional nanocomplex, termed M2Exo@CuS-CitP-Rapa (M2CPR), with the aim of selectively inhibiting inflammatory immune reactions while promoting immune tolerance towards specific antigens. M2CPR specifically targets inflammatory tissues in RA, delivering CuS NPs, CitP, Rapa, and endogenous anti-inflammatory factors, thereby ameliorating the inflammatory joint microenvironment. CuS NPs induce Cuproptosis of activated T cells, whose fragments are engulfed by resident or recruited macrophages, resulting in abundant production of TGF-β. TGF-β acts synergistically with Rapa to induce the iDCs into tDCs. tDCs present CitP to Naive T cells, promoting Tregs differentiation. Tregs, in turn, produce more TGF-β, inducing tDCs differentiation, thereby establishing a cycle of immune tolerance. Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA therapy.
用于免疫调节的纳米药物在治疗类风湿性关节炎(RA)方面取得了进展。然而,由于患者免疫系统的畸变,在诱导抗原特异性免疫耐受的同时阻止疾病进展仍是一项重大挑战。在这里,我们开发了一种高度靶向性的多功能纳米复合物,称为 M2Exo@CuS-CitP-Rapa(M2CPR),旨在选择性地抑制炎症性免疫反应,同时促进对特定抗原的免疫耐受。M2CPR 专门针对 RA 中的炎症组织,输送 CuS NPs、CitP、Rapa 和内源性抗炎因子,从而改善炎性关节微环境。CuS NPs 能诱导活化的 T 细胞发生杯突分裂,其碎片被常驻或招募的巨噬细胞吞噬,从而产生大量的 TGF-β。TGF-β 与 Rapa 协同作用,将 iDCs 诱导为 tDCs。Tregs反过来会产生更多的TGF-β,诱导tDCs分化,从而建立免疫耐受循环。通过体外和体内实验,我们验证了 M2CPR 可以诱导稳健持久的抗原特异性免疫耐受,为 RA 治疗提供了一种新的范例。
{"title":"Engineering M2 macrophage-derived exosomes modulate activated T cell cuproptosis to promote immune tolerance in rheumatoid arthritis","authors":"Guoquan Wu ,&nbsp;Tianyu Su ,&nbsp;Peng Zhou ,&nbsp;Rongze Tang ,&nbsp;Xu Zhu ,&nbsp;Jin Wang ,&nbsp;Minghao Chao ,&nbsp;Liying Fan ,&nbsp;Hanrong Yan ,&nbsp;Peng Ye ,&nbsp;Dehong Yu ,&nbsp;Fenglei Gao ,&nbsp;Hongliang Chen","doi":"10.1016/j.biomaterials.2024.122943","DOIUrl":"10.1016/j.biomaterials.2024.122943","url":null,"abstract":"<div><div>Nanomedicines for immune modulation have made advancements in the treatment of rheumatoid arthritis (RA). However, due to aberrations in patients' immune systems, inducing antigen-specific immune tolerance while halting disease progression remains a significant challenge. Here, we develop a highly targeted multifunctional nanocomplex, termed M2Exo@CuS-CitP-Rapa (M2CPR), with the aim of selectively inhibiting inflammatory immune reactions while promoting immune tolerance towards specific antigens. M2CPR specifically targets inflammatory tissues in RA, delivering CuS NPs, CitP, Rapa, and endogenous anti-inflammatory factors, thereby ameliorating the inflammatory joint microenvironment. CuS NPs induce Cuproptosis of activated T cells, whose fragments are engulfed by resident or recruited macrophages, resulting in abundant production of TGF-β. TGF-β acts synergistically with Rapa to induce the iDCs into tDCs. tDCs present CitP to Naive T cells, promoting Tregs differentiation. Tregs, in turn, produce more TGF-β, inducing tDCs differentiation, thereby establishing a cycle of immune tolerance. Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA therapy.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122943"},"PeriodicalIF":12.8,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor vascular occlusion by calcium-based thermosensitizer provokes continuous cavitation effect and thermal energy transition efficiency of radiofrequency ablation therapy 钙基热敏剂对肿瘤血管的阻塞引发持续空化效应,提高射频消融疗法的热能转换效率
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-02 DOI: 10.1016/j.biomaterials.2024.122944
Guichun Zeng , Xiayi Liang , Yuan Ling , Xiaoqi Zhu , Qin Wang , Zelun Li , Junjie Liu , Xiaobo Wang , Guanhua Qiu , Kangning Yan , Duo Wang , Jie Chen
Radiofrequency ablation (RFA) therapy for hepatocellular carcinoma (HCC) suffers from incomplete ablation with tumor remnants, recurrence, and metastasis. To capture these matters, a calcium-based thermosensitizer (CBT) was constructed, which can swell the thermal ablation treatment. DMXAA was encapsulated within CaCO3 nanoparticles and surface-modified using PEG. DMXAA @CBTNps emanates continuous cavitation to enhance the RFA effect, lower RFA power, and shorten the RFA time by responding to the acidic tumor microenvironment and releasing carbon dioxide bubbles. Ca2+ deposition to form calcification instigates the calcium death of the tumor and strengthens the thermal conductivity, wherein CBT fortifies the immunogenic cell death (ICD) of RFA. The vascular disruptor DMXAA is administered to the tumor site to impair the blood and nutrient supply to the tumor tissue. Calcium carbonate nanoparticles generate persistent carbon dioxide bubbles within the acidic microenvironment, leading to a sustained cavitation effect that enhances magneto-thermal conversion. This synergistic approach facilitates tumor vascular occlusion, thereby improving thermal ablation therapy. This strategy is different from previous thermal ablation treatments in that the CBT-released product Ca2+, the continuous cavitation effect of CO2, and the vascular disrupting agent can accelerate the conversion of energy from electromagnetic energy to thermal energy and reduce the heat loss, which significantly amplifies the effect of thermal ablation treatment of HCC and intensifies ICD. Therefore, this research provides a promising avenue and therapeutic platform for clinical liver cancer treatment.
肝细胞癌(HCC)的射频消融(RFA)治疗存在肿瘤残余消融不彻底、复发和转移等问题。为了解决这些问题,研究人员构建了一种钙基热敏剂(CBT),它可以膨胀热消融治疗。DMXAA 被封装在 CaCO3 纳米粒子中,并使用 PEG 进行表面修饰。DMXAA @CBTNps 通过响应酸性肿瘤微环境并释放二氧化碳气泡,产生持续的空化作用,从而增强 RFA 效果,降低 RFA 功率,缩短 RFA 时间。Ca2+ 沉积形成的钙化促进了肿瘤的钙化死亡并增强了导热性,而 CBT 则强化了 RFA 的免疫性细胞死亡(ICD)。在肿瘤部位施用血管破坏剂 DMXAA,以破坏肿瘤组织的血液和营养供应。碳酸钙纳米粒子在酸性微环境中产生持久的二氧化碳气泡,从而产生持续的空化效应,增强磁热转换。这种协同方法可促进肿瘤血管闭塞,从而改善热消融治疗。这种策略与以往的热消融治疗不同,CBT 释放的产物 Ca2+、二氧化碳的持续空化效应和血管破坏剂可以加速能量从电磁能到热能的转换,减少热量损失,从而显著放大 HCC 热消融治疗的效果,强化 ICD。因此,这项研究为临床肝癌治疗提供了一个前景广阔的途径和治疗平台。
{"title":"Tumor vascular occlusion by calcium-based thermosensitizer provokes continuous cavitation effect and thermal energy transition efficiency of radiofrequency ablation therapy","authors":"Guichun Zeng ,&nbsp;Xiayi Liang ,&nbsp;Yuan Ling ,&nbsp;Xiaoqi Zhu ,&nbsp;Qin Wang ,&nbsp;Zelun Li ,&nbsp;Junjie Liu ,&nbsp;Xiaobo Wang ,&nbsp;Guanhua Qiu ,&nbsp;Kangning Yan ,&nbsp;Duo Wang ,&nbsp;Jie Chen","doi":"10.1016/j.biomaterials.2024.122944","DOIUrl":"10.1016/j.biomaterials.2024.122944","url":null,"abstract":"<div><div>Radiofrequency ablation (RFA) therapy for hepatocellular carcinoma (HCC) suffers from incomplete ablation with tumor remnants, recurrence, and metastasis. To capture these matters, a calcium-based thermosensitizer (CBT) was constructed, which can swell the thermal ablation treatment. DMXAA was encapsulated within CaCO<sub>3</sub> nanoparticles and surface-modified using PEG. DMXAA @CBTNps emanates continuous cavitation to enhance the RFA effect, lower RFA power, and shorten the RFA time by responding to the acidic tumor microenvironment and releasing carbon dioxide bubbles. Ca<sup>2+</sup> deposition to form calcification instigates the calcium death of the tumor and strengthens the thermal conductivity, wherein CBT fortifies the immunogenic cell death (ICD) of RFA. The vascular disruptor DMXAA is administered to the tumor site to impair the blood and nutrient supply to the tumor tissue. Calcium carbonate nanoparticles generate persistent carbon dioxide bubbles within the acidic microenvironment, leading to a sustained cavitation effect that enhances magneto-thermal conversion. This synergistic approach facilitates tumor vascular occlusion, thereby improving thermal ablation therapy. This strategy is different from previous thermal ablation treatments in that the CBT-released product Ca<sup>2+</sup>, the continuous cavitation effect of CO<sub>2</sub>, and the vascular disrupting agent can accelerate the conversion of energy from electromagnetic energy to thermal energy and reduce the heat loss, which significantly amplifies the effect of thermal ablation treatment of HCC and intensifies ICD. Therefore, this research provides a promising avenue and therapeutic platform for clinical liver cancer treatment.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122944"},"PeriodicalIF":12.8,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142577889","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reprogramming metabolic microenvironment for nerve regeneration via waterborne polylactic acid-polyurethane copolymer scaffolds 通过水性聚乳酸-聚氨酯共聚物支架重编程神经再生代谢微环境。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-01 DOI: 10.1016/j.biomaterials.2024.122942
Yuan Feng , Jinlin Chen , Xiao Wang , Chao Long , Wenbo Wang , Jingjing Lin , Yuanyuan He , Yanchao Wang , Feng Luo , Zhen Li , Jiehua Li , Hong Tan
Cell metabolism, as the key driver of inflammation, revascularization and even subsequent tissue regeneration, is controlled by and also conversely influenced by signal transduction. Incorporation of cell metabolism into tissue engineering research holds immense potential for in-situ treatment repair and further understanding of the host-biomaterial cues in body response. In this study, an anti-inflammatory waterborne polyurethane scaffold incorporated with poly-l-lactic acid (PLLA) block was served to repair nerve injuries (LAx-WPU). Lactate was released through the degradation of LAx-WPU scaffolds, and the content increased with the addition of PLLA block over the degradation times. Thenceforth, the production of adenosine triphosphate (ATP) in primary neurons and neuronal axon growth were achieved by taking up lactate through monocarboxylate transporters (MCT2) for energy metabolism under glucose-free environment treated with LAx-WPU degradation solution. After LAx-WPU was implanted to repair brain nerve defects in rats, filamentous neurons elongation, rapid vascularization, and nerve tissue regeneration were realized up to 28 days with the positive expression of microtubule-associated protein (MAP2), β-tubulin (Tuj1), and platelet endothelial cell adhesion molecule (CD31) in the scaffolds. Results highlighted that the LAx-WPU scaffolds up-regulated not only the ATP-ADP-AMP purine metabolism compounds to mainly bridge neuroactive ligand-receptor interaction genes, cAMP pathway genes, and calcium pathway genes for neurocytes but also the ATP-GMP purine metabolism to angiogenesis in Gene Ontology (GO) analysis. Further analysis in reverse showed axonal regeneration is restrained by the inhibition of MCT2, proving LAx-WPU promoted nerve repair depended on lactate for energy. Therefore, LAx-WPU scaffolds construct an expected way to modulate the metabolic microenvironment for inducing nerve regeneration by intrinsic biomaterial metabolism cues without any bioactive factors.
细胞新陈代谢是炎症、血管再造乃至后续组织再生的关键驱动因素,它既受信号转导的控制,也反过来受信号转导的影响。将细胞新陈代谢纳入组织工程研究,对于原位治疗修复和进一步了解宿主-生物材料在机体反应中的线索具有巨大潜力。在这项研究中,一种含有聚乳酸块的抗炎水性聚氨酯支架被用于修复神经损伤(LAx-WPU)。LAx-WPU 支架在降解过程中会释放出乳酸,随着聚乳酸块的加入,乳酸的含量会随着降解时间的延长而增加。此后,在经 LAx-WPU 降解液处理的无葡萄糖环境中,原代神经元通过单羧酸盐转运体(MCT2)吸收乳酸进行能量代谢,从而产生三磷酸腺苷(ATP)并促进神经轴突生长。将 LAx-WPU 植入大鼠脑部神经缺损修复后,28 天内大鼠脑部神经组织实现了丝状神经元伸长、快速血管化和神经组织再生,支架上的微管相关蛋白(MAP2)、β-微管蛋白(Tuj1)和血小板内皮细胞粘附分子(CD31)均呈阳性表达。结果表明,LAx-WPU支架不仅上调了ATP-ADP-AMP嘌呤代谢化合物与神经细胞的神经活性配体-受体相互作用基因、cAMP通路基因和钙通路基因之间的主要桥梁作用,而且在基因本体(GO)分析中还上调了ATP-GMP嘌呤代谢与血管生成之间的关系。进一步的反向分析表明,轴突再生受到 MCT2 的抑制,证明 LAx-WPU 促进神经修复依赖乳酸提供能量。因此,LAx-WPU支架有望在不使用任何生物活性因子的情况下,通过内在的生物材料代谢线索来调节代谢微环境,从而诱导神经再生。
{"title":"Reprogramming metabolic microenvironment for nerve regeneration via waterborne polylactic acid-polyurethane copolymer scaffolds","authors":"Yuan Feng ,&nbsp;Jinlin Chen ,&nbsp;Xiao Wang ,&nbsp;Chao Long ,&nbsp;Wenbo Wang ,&nbsp;Jingjing Lin ,&nbsp;Yuanyuan He ,&nbsp;Yanchao Wang ,&nbsp;Feng Luo ,&nbsp;Zhen Li ,&nbsp;Jiehua Li ,&nbsp;Hong Tan","doi":"10.1016/j.biomaterials.2024.122942","DOIUrl":"10.1016/j.biomaterials.2024.122942","url":null,"abstract":"<div><div>Cell metabolism, as the key driver of inflammation, revascularization and even subsequent tissue regeneration, is controlled by and also conversely influenced by signal transduction. Incorporation of cell metabolism into tissue engineering research holds immense potential for in-situ treatment repair and further understanding of the host-biomaterial cues in body response. In this study, an anti-inflammatory waterborne polyurethane scaffold incorporated with poly-<span>l</span>-lactic acid (PLLA) block was served to repair nerve injuries (LAx-WPU). Lactate was released through the degradation of LAx-WPU scaffolds, and the content increased with the addition of PLLA block over the degradation times. Thenceforth, the production of adenosine triphosphate (ATP) in primary neurons and neuronal axon growth were achieved by taking up lactate through monocarboxylate transporters (MCT2) for energy metabolism under glucose-free environment treated with LAx-WPU degradation solution. After LAx-WPU was implanted to repair brain nerve defects in rats, filamentous neurons elongation, rapid vascularization, and nerve tissue regeneration were realized up to 28 days with the positive expression of microtubule-associated protein (MAP2), β-tubulin (Tuj1), and platelet endothelial cell adhesion molecule (CD31) in the scaffolds. Results highlighted that the LAx-WPU scaffolds up-regulated not only the ATP-ADP-AMP purine metabolism compounds to mainly bridge neuroactive ligand-receptor interaction genes, cAMP pathway genes, and calcium pathway genes for neurocytes but also the ATP-GMP purine metabolism to angiogenesis in Gene Ontology (GO) analysis. Further analysis in reverse showed axonal regeneration is restrained by the inhibition of MCT2, proving LAx-WPU promoted nerve repair depended on lactate for energy. Therefore, LAx-WPU scaffolds construct an expected way to modulate the metabolic microenvironment for inducing nerve regeneration by intrinsic biomaterial metabolism cues without any bioactive factors.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122942"},"PeriodicalIF":12.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Implantation of MSC spheroid-derived 3D decellularized ECM enriched with the MSC secretome ameliorates traumatic brain injury and promotes brain repair 植入富含间充质干细胞分泌组的间充质干细胞球衍生三维脱细胞ECM可改善创伤性脑损伤并促进脑修复。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-11-01 DOI: 10.1016/j.biomaterials.2024.122941
Grace H. Chen , Kee-Chin Sia , Shao-Wen Liu , Ying-Chi Kao , Pei-Ching Yang , Chia-Hsin Ho , Shih-Chen Huang , Peng-Ying Lee , Min-Zong Liang , Linyi Chen , Chieh-Cheng Huang
Traumatic brain injury (TBI) presents substantial clinical challenges, as existing treatments are unable to reverse damage or effectively promote brain tissue regeneration. Although implantable biomaterials have been proposed to support tissue repair by mitigating the adverse microenvironment in injured brains, many fail to replicate the complex composition and architecture of the native extracellular matrix (ECM), resulting in only limited therapeutic outcomes. This study introduces an innovative approach by developing a mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) decellularized ECM (dECM) that is enriched with the MSC-derived matrisome and secretome, offering a promising solution for TBI treatment and brain tissue regeneration. Proteomic and cytokine array analyses revealed that 3D dECM retained a diverse array of MSC spheroid-derived matrisome proteins and secretome components, which are crucial for replicating the complexity of native ECM and the therapeutic capabilities of MSCs. These molecules were found to underlie the observed effects of 3D dECM on immunomodulation, proneuritogenesis, and proangiogenesis in our in vitro functional assays. Implantation of 3D dECM into TBI model mice effectively mitigated postinjury tissue damage and promoted brain repair, as evidenced by a reduced brain lesion volume, decreased cell apoptosis, alleviated neuroinflammation, reduced glial scar formation, and increased of neuroblast recruitment to the lesion site. These outcomes culminated in improved motor function recovery in animals, highlighting the multifaceted therapeutic potential of 3D dECM for TBI. In summary, our study elucidates the transformative potential of MSC spheroid-derived bioactive 3D dECM as an implantable biomaterial for effectively mitigating post-TBI neurological damage, paving the way for its broader therapeutic application.
创伤性脑损伤(TBI)给临床带来了巨大挑战,因为现有的治疗方法无法逆转损伤或有效促进脑组织再生。虽然有人提出植入式生物材料可通过缓解受伤大脑中的不利微环境来支持组织修复,但许多生物材料无法复制原生细胞外基质(ECM)的复杂成分和结构,导致治疗效果有限。本研究引入了一种创新方法,即开发一种间充质干细胞(MSC)球体衍生的三维(3D)脱细胞ECM(dECM),该ECM富含间充质干细胞衍生的基质组(MSC-derived matrisome)和分泌组(Secretome),为创伤性脑损伤治疗和脑组织再生提供了一种前景广阔的解决方案。蛋白质组和细胞因子阵列分析表明,三维 dECM 保留了多种间充质干细胞球衍生的基质组蛋白和分泌组成分,这对于复制原生 ECM 的复杂性和间充质干细胞的治疗能力至关重要。在我们的体外功能测试中,我们发现这些分子是三维 dECM 对免疫调节、促泌尿素生成和促血管生成的观察效应的基础。将三维 dECM 植入创伤性脑损伤模型小鼠体内可有效减轻伤后组织损伤并促进脑修复,具体表现为脑损伤体积缩小、细胞凋亡减少、神经炎症减轻、胶质瘢痕形成减少以及病变部位神经母细胞募集增加。这些结果最终改善了动物的运动功能恢复,凸显了三维 dECM 对创伤性脑损伤的多方面治疗潜力。总之,我们的研究阐明了间充质干细胞球体衍生的生物活性三维 dECM 作为一种可植入的生物材料在有效缓解创伤后神经损伤方面的变革潜力,为其更广泛的治疗应用铺平了道路。
{"title":"Implantation of MSC spheroid-derived 3D decellularized ECM enriched with the MSC secretome ameliorates traumatic brain injury and promotes brain repair","authors":"Grace H. Chen ,&nbsp;Kee-Chin Sia ,&nbsp;Shao-Wen Liu ,&nbsp;Ying-Chi Kao ,&nbsp;Pei-Ching Yang ,&nbsp;Chia-Hsin Ho ,&nbsp;Shih-Chen Huang ,&nbsp;Peng-Ying Lee ,&nbsp;Min-Zong Liang ,&nbsp;Linyi Chen ,&nbsp;Chieh-Cheng Huang","doi":"10.1016/j.biomaterials.2024.122941","DOIUrl":"10.1016/j.biomaterials.2024.122941","url":null,"abstract":"<div><div>Traumatic brain injury (TBI) presents substantial clinical challenges, as existing treatments are unable to reverse damage or effectively promote brain tissue regeneration. Although implantable biomaterials have been proposed to support tissue repair by mitigating the adverse microenvironment in injured brains, many fail to replicate the complex composition and architecture of the native extracellular matrix (ECM), resulting in only limited therapeutic outcomes. This study introduces an innovative approach by developing a mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) decellularized ECM (dECM) that is enriched with the MSC-derived matrisome and secretome, offering a promising solution for TBI treatment and brain tissue regeneration. Proteomic and cytokine array analyses revealed that 3D dECM retained a diverse array of MSC spheroid-derived matrisome proteins and secretome components, which are crucial for replicating the complexity of native ECM and the therapeutic capabilities of MSCs. These molecules were found to underlie the observed effects of 3D dECM on immunomodulation, proneuritogenesis, and proangiogenesis in our <em>in vitro</em> functional assays. Implantation of 3D dECM into TBI model mice effectively mitigated postinjury tissue damage and promoted brain repair, as evidenced by a reduced brain lesion volume, decreased cell apoptosis, alleviated neuroinflammation, reduced glial scar formation, and increased of neuroblast recruitment to the lesion site. These outcomes culminated in improved motor function recovery in animals, highlighting the multifaceted therapeutic potential of 3D dECM for TBI. In summary, our study elucidates the transformative potential of MSC spheroid-derived bioactive 3D dECM as an implantable biomaterial for effectively mitigating post-TBI neurological damage, paving the way for its broader therapeutic application.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122941"},"PeriodicalIF":12.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602159","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ru single-atom nanozymes targeting ROS-ferroptosis pathways for enhanced endometrial regeneration in intrauterine adhesion therapy 针对 ROS-ferroptosis 通路的 Ru 单原子纳米酶,在宫内粘连治疗中促进子宫内膜再生。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-10-30 DOI: 10.1016/j.biomaterials.2024.122923
Yuxiang Liang , Jian Meng , Zhaowei Yu , Yuqian Guo , Xiao Zhang , Yujia Yan , Shaobo Du , Shanshan Jin , Jing Li , Hailan Yang , Xiaozheng Zhang , Zhizhen Liu , Liping Li , Jun Xie
Intrauterine adhesion (IUA) presents a significant challenge in gynecology, characterized by excessive fibrosis and compromised reproductive function, leading to severe infertility. Although biocompatible hydrogels integrated with stem cells offer a promising approach for IUA therapy, clinical applications remain limited. Recent studies have highlighted the role of ferroptosis and reactive oxygen species (ROS) in IUA pathogenesis, yet strategies targeting ferroptosis through antioxidant stress are underexplored. This study investigates the therapeutic effects and mechanisms of a Ru-Single-Atom Nanozyme (Ru-SAN) incorporated into chitosan hydrogel for treating IUA. Ru-SAN, which mimics the enzyme activities of catalase, superoxide dismutase, and glutathione peroxidase, effectively clears excess ROS and shows promise in treating oxidative stress-induced diseases. The results demonstrate the superior antioxidative capabilities of Ru-SAN, significantly suppressing the ROS-ferroptosis cycle at the injury site. This creates a favorable microenvironment for post-injury repair by inhibiting inflammation, enhancing mesenchymal-to-epithelial transformation, promoting angiogenesis, and polarizing M2 macrophages. Importantly, it mitigates adverse repair outcomes from inflammation and excessive collagen fiber deposition, ultimately restoring uterine glandular structures and thickness, thereby achieving the ultimate goal of restoring fertility and live birth rates. In conclusion, our study delineates a pioneering therapeutic approach leveraging the antioxidant properties of Ru-SAN to target ferroptosis, thereby offering an efficacious treatment for IUA.
宫腔内粘连(IUA)是妇科领域的一项重大挑战,其特点是过度纤维化和生殖功能受损,导致严重不孕。虽然与干细胞结合的生物相容性水凝胶为IUA治疗提供了一种前景广阔的方法,但临床应用仍然有限。最近的研究强调了铁突变和活性氧(ROS)在IUA发病机制中的作用,但通过抗氧化应激靶向铁突变的策略尚未得到充分探索。本研究探讨了加入壳聚糖水凝胶的 Ru-单原子纳米酶(Ru-SAN)治疗 IUA 的疗效和机制。Ru-SAN 可模拟过氧化氢酶、超氧化物歧化酶和谷胱甘肽过氧化物酶的酶活性,能有效清除过量的 ROS,有望治疗氧化应激诱发的疾病。研究结果表明,Ru-SAN 具有卓越的抗氧化能力,能显著抑制损伤部位的 ROS-ferroptosis 循环。这通过抑制炎症、加强间质到上皮的转化、促进血管生成和极化 M2 巨噬细胞,为损伤后的修复创造了有利的微环境。重要的是,它能减轻炎症和胶原纤维过度沉积造成的不良修复结果,最终恢复子宫腺体结构和厚度,从而实现恢复生育能力和活产率的最终目标。总之,我们的研究描述了一种开创性的治疗方法,利用 Ru-SAN 的抗氧化特性来靶向铁蛋白沉积,从而为 IUA 提供了一种有效的治疗方法。
{"title":"Ru single-atom nanozymes targeting ROS-ferroptosis pathways for enhanced endometrial regeneration in intrauterine adhesion therapy","authors":"Yuxiang Liang ,&nbsp;Jian Meng ,&nbsp;Zhaowei Yu ,&nbsp;Yuqian Guo ,&nbsp;Xiao Zhang ,&nbsp;Yujia Yan ,&nbsp;Shaobo Du ,&nbsp;Shanshan Jin ,&nbsp;Jing Li ,&nbsp;Hailan Yang ,&nbsp;Xiaozheng Zhang ,&nbsp;Zhizhen Liu ,&nbsp;Liping Li ,&nbsp;Jun Xie","doi":"10.1016/j.biomaterials.2024.122923","DOIUrl":"10.1016/j.biomaterials.2024.122923","url":null,"abstract":"<div><div>Intrauterine adhesion (IUA) presents a significant challenge in gynecology, characterized by excessive fibrosis and compromised reproductive function, leading to severe infertility. Although biocompatible hydrogels integrated with stem cells offer a promising approach for IUA therapy, clinical applications remain limited. Recent studies have highlighted the role of ferroptosis and reactive oxygen species (ROS) in IUA pathogenesis, yet strategies targeting ferroptosis through antioxidant stress are underexplored. This study investigates the therapeutic effects and mechanisms of a Ru-Single-Atom Nanozyme (Ru-SAN) incorporated into chitosan hydrogel for treating IUA. Ru-SAN, which mimics the enzyme activities of catalase, superoxide dismutase, and glutathione peroxidase, effectively clears excess ROS and shows promise in treating oxidative stress-induced diseases. The results demonstrate the superior antioxidative capabilities of Ru-SAN, significantly suppressing the ROS-ferroptosis cycle at the injury site. This creates a favorable microenvironment for post-injury repair by inhibiting inflammation, enhancing mesenchymal-to-epithelial transformation, promoting angiogenesis, and polarizing M2 macrophages. Importantly, it mitigates adverse repair outcomes from inflammation and excessive collagen fiber deposition, ultimately restoring uterine glandular structures and thickness, thereby achieving the ultimate goal of restoring fertility and live birth rates. In conclusion, our study delineates a pioneering therapeutic approach leveraging the antioxidant properties of Ru-SAN to target ferroptosis, thereby offering an efficacious treatment for IUA.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122923"},"PeriodicalIF":12.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bimetallic peroxide-based nanotherapeutics for immunometabolic intervention and induction of immunogenic cell death to augment cancer immunotherapy 基于双金属过氧化物的纳米疗法,用于免疫代谢干预和诱导免疫原性细胞死亡,以增强癌症免疫疗法。
IF 12.8 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-10-30 DOI: 10.1016/j.biomaterials.2024.122934
Min Han , Shiying Zhou , Zunde Liao , Chen Zishan , Xiangting Yi , Chuanbin Wu , Dongmei Zhang , Yao He , Kam W. Leong , Yiling Zhong
Immunotherapy has transformed cancer treatment, but its efficacy is often limited by the immunosuppressive characteristics of the tumor microenvironment (TME), which are predominantly influenced by the metabolism of cancer cells. Among these metabolic pathways, the indoleamine 2,3-dioxygenase (IDO) pathway is particularly crucial, as it significantly contributes to TME suppression and influences immune cell activity. Additionally, inducing immunogenic cell death (ICD) in tumor cells can reverse the immunosuppressive TME, thereby enhancing the efficacy of immunotherapy. Herein, we develop CGDMRR, a novel bimetallic peroxide-based nanodrug based on copper-cerium peroxide nanoparticles. These nanotherapeutics are engineered to mitigate tumor hypoxia and deliver therapeutics such as 1-methyltryptophan (1MT), glucose oxidase (GOx), and doxorubicin (Dox) in a targeted manner. The design aims to alleviate tumor hypoxia, reduce the immunosuppressive effects of the IDO pathway, and promote ICD. CGDMRR effectively inhibits the growth of 4T1 tumors and elicits antitumor immune responses by leveraging immunometabolic interventions and therapies that induce ICD. Furthermore, when CGDMRR is combined with a clinically certified anti-PD-L1 antibody, its efficacy in inhibiting tumor growth is enhanced. This improved efficacy extends beyond unilateral tumor models, also affecting bilateral tumors and lung metastases, due to the activation of systemic antitumor immunity. This study underscores CGDMRR's potential to augment the efficacy of PD-L1 blockade in breast cancer immunotherapy.
免疫疗法改变了癌症治疗,但其疗效往往受到肿瘤微环境(TME)免疫抑制特性的限制,而肿瘤微环境主要受癌细胞代谢的影响。在这些代谢途径中,吲哚胺-2,3-二氧化酶(IDO)途径尤为重要,因为它在很大程度上促进了肿瘤微环境的抑制并影响了免疫细胞的活性。此外,诱导肿瘤细胞的免疫原性细胞死亡(ICD)可以逆转免疫抑制的TME,从而提高免疫疗法的疗效。在此,我们开发了一种基于过氧化铜铈纳米颗粒的新型双金属过氧化物基纳米药物 CGDMRR。这些纳米治疗药物可缓解肿瘤缺氧,并以靶向方式递送治疗药物,如 1-甲基色氨酸(1MT)、葡萄糖氧化酶(GOx)和多柔比星(Dox)。该设计旨在缓解肿瘤缺氧,减少 IDO 通路的免疫抑制作用,促进 ICD。CGDMRR 可有效抑制 4T1 肿瘤的生长,并利用免疫代谢干预和诱导 ICD 的疗法激发抗肿瘤免疫反应。此外,当 CGDMRR 与临床认证的抗 PD-L1 抗体结合使用时,其抑制肿瘤生长的功效会得到增强。由于激活了全身性的抗肿瘤免疫,这种疗效的提高超越了单侧肿瘤模型,也影响到双侧肿瘤和肺转移瘤。这项研究强调了 CGDMRR 在乳腺癌免疫疗法中增强 PD-L1 阻断疗效的潜力。
{"title":"Bimetallic peroxide-based nanotherapeutics for immunometabolic intervention and induction of immunogenic cell death to augment cancer immunotherapy","authors":"Min Han ,&nbsp;Shiying Zhou ,&nbsp;Zunde Liao ,&nbsp;Chen Zishan ,&nbsp;Xiangting Yi ,&nbsp;Chuanbin Wu ,&nbsp;Dongmei Zhang ,&nbsp;Yao He ,&nbsp;Kam W. Leong ,&nbsp;Yiling Zhong","doi":"10.1016/j.biomaterials.2024.122934","DOIUrl":"10.1016/j.biomaterials.2024.122934","url":null,"abstract":"<div><div>Immunotherapy has transformed cancer treatment, but its efficacy is often limited by the immunosuppressive characteristics of the tumor microenvironment (TME), which are predominantly influenced by the metabolism of cancer cells. Among these metabolic pathways, the indoleamine 2,3-dioxygenase (IDO) pathway is particularly crucial, as it significantly contributes to TME suppression and influences immune cell activity. Additionally, inducing immunogenic cell death (ICD) in tumor cells can reverse the immunosuppressive TME, thereby enhancing the efficacy of immunotherapy. Herein, we develop CGDMRR, a novel bimetallic peroxide-based nanodrug based on copper-cerium peroxide nanoparticles. These nanotherapeutics are engineered to mitigate tumor hypoxia and deliver therapeutics such as 1-methyltryptophan (1MT), glucose oxidase (GOx), and doxorubicin (Dox) in a targeted manner. The design aims to alleviate tumor hypoxia, reduce the immunosuppressive effects of the IDO pathway, and promote ICD. CGDMRR effectively inhibits the growth of 4T1 tumors and elicits antitumor immune responses by leveraging immunometabolic interventions and therapies that induce ICD. Furthermore, when CGDMRR is combined with a clinically certified anti-PD-L1 antibody, its efficacy in inhibiting tumor growth is enhanced. This improved efficacy extends beyond unilateral tumor models, also affecting bilateral tumors and lung metastases, due to the activation of systemic antitumor immunity. This study underscores CGDMRR's potential to augment the efficacy of PD-L1 blockade in breast cancer immunotherapy.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"315 ","pages":"Article 122934"},"PeriodicalIF":12.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biomaterials
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