Decellularized Extracellular Matrix

脱细胞细胞外基质
  • 文章类型: Journal Article
    在组织工程领域,细胞外基质(ECM)被认为是促进脊髓损伤(SCI)后神经再生的重要元素。牙髓干细胞(DPSC),源自神经c的间充质干细胞,易于在体外收获和培养,表达多种神经营养因子(NTFs)并沉积大量ECM,使它们成为干细胞或基于ECM的SCI治疗的好选择。在本研究中,衍生自DPSC片的脱细胞细胞外基质(dECM)用于治疗SCI。优化实验表明,将DPSC片与1%TritonX-100孵育5分钟是制备DPSCdECM的最佳程序。发现DPSCdECM促进SCI后神经修复和再生,并恢复大鼠后肢运动功能。机械上,DPSCdECM促进神经干细胞的迁移和神经分化,以及小胶质细胞的M2极化,抑制胶质疤痕的形成.这项研究表明,使用DPSCdECM是治疗SCI的潜在策略。
    In the field of tissue engineering, the extracellular matrix (ECM) is considered an important element for promoting neural regeneration after spinal cord injury (SCI). Dental pulp stem cells (DPSCs), mesenchymal stem cells that originate from the neural crest, are easy to harvest and culture in vitro, express a variety of neurotrophic factors (NTFs) and deposit a large amount of ECM, making them a good choice for stem cell- or ECM-based treatment of SCI. In the present study, decellularized extracellular matrix (dECM) derived from DPSC sheets is used for the treatment of SCI. Optimization experiments reveal that incubating DPSC sheets with 1% Triton X-100 for 5 min is the best procedure for preparing DPSC dECM. It is found that DPSC dECM promotes nerve repair and regeneration after SCI and restores hindlimb motor function in rats. Mechanistically, DPSC dECM facilitates the migration and neural differentiation of neural stem cells, as well as M2 polarization of microglia, and inhibits the formation of glial scars. This study suggests that the use of DPSC dECM is a potential strategy for the treatment of SCI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肺纤维化(PF)是由成纤维细胞活化为肌成纤维细胞后的过度细胞外基质(ECM)沉积和组织重塑引起的。异常沉积的纤维化ECM,反过来,促进成纤维细胞活化并加速肺结构和功能的丧失。然而,纤维化ECM促进成纤维细胞活化的分子介质和确切机制尚不清楚.在博来霉素诱导的PF小鼠模型中,我们发现Galectin-1(Gal-1)在肺组织中的表达显著增加,和Gal-1质粒转染的成纤维细胞的过表达被激活为肌成纤维细胞。利用脱细胞技术制备脱细胞纤维化ECM,并与成纤维细胞构建3D体外共培养体系,我们发现去细胞纤维化ECM诱导Gal-1的高表达并促进成纤维细胞活化为肌成纤维细胞。因此,Gal-1已被确定为PF中的关键介体。Further,我们发现去细胞纤维化ECM通过Gal-1介导的FAK-Src-P130Cas机械信号通路向细胞传递机械信号,而CYP450酶(主要涉及CYP1A1,CYP24A1,CYP3A4和CYP2D6亚型)充当化学信号通路,以接收从上游Gal-1传递的机械信号,从而促进成纤维细胞活化。Gal-1抑制剂OTX008或CYP1A1抑制剂7-羟基黄酮在小鼠中预防PF并抑制纤维化ECM在促进成纤维细胞活化为肌成纤维细胞中的作用,防止PF。这些结果揭示了肺纤维化形成的新分子机制,并鉴定了Gal-1及其下游CYP1A1作为PF疾病治疗的潜在治疗靶标。
    Pulmonary fibrosis (PF) results from excessive extracellular matrix (ECM) deposition and tissue remodeling after activation of fibroblasts into myofibroblasts. Abnormally deposited fibrotic ECM, in turn, promotes fibroblast activation and accelerates loss of lung structure and function. However, the molecular mediators and exact mechanisms by which fibrotic ECM promotes fibroblast activation are unclear. In a bleomycin-induced PF mouse model, we found Galectin-1 (Gal-1) expression was significantly increased in lung tissue, and overexpression of Gal-1 plasmid-transfected fibroblasts were activated into myofibroblasts. Using the decellularization technique to prepare decellularized fibrotic ECM and constructing a 3D in vitro co-culture system with fibroblasts, we found that decellularized fibrotic ECM induced a high expression of Gal-1 and promoted the activation of fibroblasts into myofibroblasts. Therefore, Gal-1 has been identified as a pivotal mediator in PF. Further, we found that decellularized fibrotic ECM delivered mechanical signals to cells through the Gal-1-mediated FAK-Src-P130Cas mechanical signalling pathway, while the CYP450 enzymes (mainly involved in CYP1A1, CYP24A1, CYP3A4, and CYP2D6 isoforms) acted as a chemical signalling pathway to receive mechanical signals transmitted from upstream Gal-1, thereby promoting fibroblast activation. The Gal-1 inhibitor OTX008 or the CYP1A1 inhibitor 7-Hydroxyflavone prevented PF in mice and inhibited the role of fibrotic ECM in promoting fibroblast activation into myofibroblasts, preventing PF. These results reveal novel molecular mechanisms of lung fibrosis formation and identify Gal-1 and its downstream CYP1A1 as potential therapeutic targets for PF disease treatmnts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    已知间充质基质/干细胞(MSC)及其分泌组在许多病理状态下发挥有益作用。然而,由于不合适的体外维持条件,MSC的治疗性质可能降低。标准培养方案忽略了这样一个事实,即MSC在体内存在与细胞外基质(ECM)最紧密的联系,复杂的蛋白质网络提供了一个有指导意义的微环境。我们最近发现,在细胞来源的脱细胞细胞外基质(CM-dECM)上培养的人子宫内膜MSCs的条件培养基中大量富含一些旁分泌因子,如GM-CSF,FGF-2,HGF,MMP-1,MCP-1,IL-6,IL-8,CXCL-1,-2,-5,-6(Ushakov等人。,2024).鉴于几种上调的分子属于已知参与骨骼肌再生的肌细胞,我们假设CM-dECM可能促进受损肌肉组织的恢复。这里,我们发现CM-dECM注射到氯化钡损伤的小鼠胫骨前肌中,导致肌纤维肥大并促进血管生成。此外,CM-dECM显着促进鼠C2C12成肌细胞细胞周期的进展,表明体内肌肉修复可能与刺激常驻成肌细胞增殖有关。在这项研究中,首先概述了在dECM上培养的子宫内膜MSCs的分泌组在损伤的鼠骨骼肌再生中的作用。我们的发现表明dECM上的培养可以被认为是增强MSC治疗潜力的新型预处理方法。
    Mesenchymal stromal/stem cells (MSCs) and their secretome are known to exert beneficial effects in many pathological states. However, MSCs therapeutic properties can be reduced due to unsuitable in vitro maintenance conditions. Standard culture protocols neglect the fact that MSCs exist in vivo in the closest connection with the extracellular matrix (ECM), the complex protein network providing an instructive microenvironment. We found recently that conditioned medium from human endometrial MSCs cultured on cell-derived decellularized extracellular matrix (CM-dECM) is dramatically enriched in a number of paracrine factors such as GM-CSF, FGF-2, HGF, MMP-1, MCP-1, IL-6, IL-8, CXCL-1, -2, -5, -6 (Ushakov et al., 2024). Given that several upregulated molecules belong to myokines that are known to participate in skeletal muscle regeneration, we hypothesized that CM-dECM may promote restoration of damaged muscle tissue. Here, we found that CM-dECM injections into barium chloride-injured murine m. tibialis anterior caused myofiber hypertrophy and promoted angiogenesis. Besides, CM-dECM significantly contributed to progression of murine C2C12 myoblasts cell cycle suggesting that muscle repair in vivo may be connected with stimulation of resident myoblasts proliferation. In this study, a role for secretome of endometrial MSCs cultured on dECM in injured murine skeletal muscle regeneration was outlined first. Our findings demonstrate that culture on dECM may be considered as a novel preconditioning approach enhancing MSCs therapeutic potential.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    生物材料在工程化血管化组织方面的进步中的首要目标之一不仅是复制预期组织的组成,而且是为了产生更厚的结构,其包含用于充足营养和氧气供应的血管网络。第一次,就目前所知,开发了一种临床相关的生物材料,证明由人脱细胞细胞外基质制成的水凝胶可以同时表现出强大的机械性能(在kPa范围内)和血管生成能力。这些特性使得人脐静脉内皮细胞的培养和组织成为管状结构,在体外维持其完整性14天,而不需要额外的聚合物或血管生成相关因子。这是通过重新利用胎盘绒毛膜(CM)来实现的,具有特殊生化成分的医疗废物,成为生物工程目的的宝贵资源。去细胞化后,CM用甲基丙烯酰基进行了化学修饰,产生甲基丙烯酸酯化CM(CMMA)。CMMA保留的关键蛋白,以及糖胺聚糖.所得的水凝胶快速光聚合并具有增强的强度和可定制的机械性能。此外,它们在体外和体内证明了血管生成能力,作为血管化组织工程的人性化平台,具有重要的前景。
    One of the foremost targets in the advancement of biomaterials to engineer vascularized tissues is not only to replicate the composition of the intended tissue but also to create thicker structures incorporating a vascular network for adequate nutrients and oxygen supply. For the first time, to the best of current knowledge, a clinically relevant biomaterial is developed, demonstrating that hydrogels made from the human decellularized extracellular matrix can exhibit robust mechanical properties (in the kPa range) and angiogenic capabilities simultaneously. These properties enable the culture and organization of human umbilical vein endothelial cells into tubular structures, maintaining their integrity for 14 days in vitro without the need for additional polymers or angiogenesis-related factors. This is achieved by repurposing the placenta chorionic membrane (CM), a medical waste with an exceptional biochemical composition, into a valuable resource for bioengineering purposes. After decellularization, the CM underwent chemical modification with methacryloyl groups, giving rise to methacrylated CM (CMMA). CMMA preserved key proteins, as well as glycosaminoglycans. The resulting hydrogels rapidly photopolymerize and have enhanced strength and customizable mechanical properties. Furthermore, they demonstrate angio-vasculogenic competence in vitro and in vivo, holding significant promise as a humanized platform for the engineering of vascularized tissues.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    再生周围神经界面(RPNI)通过重建目标肌肉为神经损伤提供了治疗解决方案。然而,将横切的周围神经植入RPNI中的自体骨骼肌移植物会导致供体部位的发病率,突出了对组织工程骨骼肌构建体的需求。这里,使用3D骨骼细胞打印结合直接静电纺丝开发了工程化的再生孤立周围神经界面(eRIPEN),以创建用于宿主神经植入的纳米纤维膜包裹。在这项体内研究中,经过8个月的RPNI手术,eRIPEN的最小Feret直径为15-20µm,横截面积为100-500µm2,代表肌纤维的最大分布。此外,观察到神经肌肉接头形成和肌肉收缩,力≈28N。值得注意的是,在eRIPEN组中发现对机械/热刺激的超敏反应降低,胫骨功能指数从-77提高到-56.eRIPEN的新概念为组织工程构建体在RPNI中的利用和应用铺平了道路。最终通过突触连接实现神经假体控制。
    A regenerative peripheral nerve interface (RPNI) offers a therapeutic solution for nerve injury through reconstruction of the target muscle. However, implanting a transected peripheral nerve into an autologous skeletal muscle graft in RPNI causes donor-site morbidity, highlighting the need for tissue-engineered skeletal muscle constructs. Here, an engineered regenerative isolated peripheral nerve interface (eRIPEN) is developed using 3D skeletal cell printing combined with direct electrospinning to create a nanofiber membrane envelop for host nerve implantation. In this in vivo study, after over 8 months of RPNI surgery, the eRIPEN exhibits a minimum Feret diameter of 15-20 µm with a cross-sectional area of 100-500 µm2, representing the largest distribution of myofibers. Furthermore, neuromuscular junction formation and muscle contraction with a force of ≈28 N are observed. Notably, the decreased hypersensitivity to mechanical/thermal stimuli and an improved tibial functional index from -77 to -56 are found in the eRIPEN group. The present novel concept of eRIPEN paves the way for the utilization and application of tissue-engineered constructs in RPNI, ultimately realizing neuroprosthesis control through synaptic connections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心脏纤维化发生在心肌损伤后,其特征是非顺应性细胞外基质(ECM)的异常积累,这会损害心肌细胞的收缩活动,最终导致心力衰竭。这种现象是由心脏成纤维细胞(cFbs)激活为肌成纤维细胞,并导致ECM生化变化,结构和机械性能。迄今为止,缺乏预测心脏纤维化的体外模型阻碍了对创新治疗方法的探索,因为大多数基于细胞的体外还原模型没有考虑ECM线索在驱动病理进展中的主导作用。这里,我们设计了单步脱细胞方案,以获得并全面表征由人诱导多能干细胞(iPSCs)分化的活化cFbs分泌的ECM的生化和微机械特性.我们通过调节碱性成纤维细胞生长因子(bFGF)和转化生长因子β1(TGF-β1)信号,将iPSC衍生的cFbs激活为肌成纤维细胞表型,并证实激活的细胞获得了肌成纤维细胞表型的关键特征。像SMAD2/3核穿梭,形成对齐的α-平滑肌肌动蛋白(α-SMA)-丰富的应力纤维和增加的粘着斑(FAs)组装。接下来,我们用了质谱,纳米压痕,扫描电子和共聚焦显微镜来揭示丰富的特征成分和粘弹性,心肌成纤维细胞在体外沉积的富含胶原蛋白的ECM。最后,我们证明了纤维化ECM激活iPSC衍生的心肌细胞中的机械敏感性途径,影响它们的形状,sarcomere组件,表型,和钙处理性能。因此,我们提出了人类生物启发的脱细胞基质作为无动物,等基因心肌细胞培养基质概括了心脏纤维化过程中在细胞水平上发生的关键病理生理变化。
    Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the activation of cardiac fibroblasts (cFbs) to myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments, as most of the cellular-based in vitro reductionist models do not take into account the leading role of ECM cues in driving the progression of the pathology. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α-SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro. Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere assembly, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目前的工程合成支架无法功能性修复和再生破裂的天然肌腱组织,部分原因是它们不能同时满足这些组织的独特生物学和生物力学特性。肌腱修复和再生的理想支架需要提供多孔的地形结构和生物线索,这是嵌入干细胞有效浸润和肌腱分化所必需的。为了获得卷曲和多孔的支架,高度对齐的聚(l-丙交酯)纤维是通过静电纺丝,然后进行后处理制备的。通过温和可控的氢气发泡技术,我们成功地将卷曲的纤维垫转化为三维多孔支架,而不牺牲卷曲的微观结构。然后通过纤维表面改性和碳二亚胺化学将猪衍生的脱细胞肌腱基质接枝到该多孔支架上。这些生物功能化的,卷曲,多孔支架支持了增殖,迁移,和肌腱源性干/祖细胞的肌腱诱导,同时能够粘附到天然肌腱。一起,我们的数据表明,这些生物功能化的支架可以作为有前途的工程支架用于治疗急性肌腱断裂。
    Current engineered synthetic scaffolds fail to functionally repair and regenerate ruptured native tendon tissues, partly because they cannot satisfy both the unique biological and biomechanical properties of these tissues. Ideal scaffolds for tendon repair and regeneration need to provide porous topographic structures and biological cues necessary for the efficient infiltration and tenogenic differentiation of embedded stem cells. To obtain crimped and porous scaffolds, highly aligned poly(l-lactide) fibers were prepared by electrospinning followed by postprocessing. Through a mild and controlled hydrogen gas foaming technique, we successfully transformed the crimped fibrous mats into three-dimensional porous scaffolds without sacrificing the crimped microstructure. Porcine derived decellularized tendon matrix was then grafted onto this porous scaffold through fiber surface modification and carbodiimide chemistry. These biofunctionalized, crimped, and porous scaffolds supported the proliferation, migration, and tenogenic induction of tendon derived stem/progenitor cells, while enabling adhesion to native tendons. Together, our data suggest that these biofunctionalized scaffolds can be exploited as promising engineered scaffolds for the treatment of acute tendon rupture.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    器官和组织功能障碍代表临床上显著的病症。通过将细胞生物学与材料科学相结合,组织工程能够重建和恢复受损的组织或器官,提供非侵入性修复方法。在我们的研究中,我们通过利用人脐带衍生的脱细胞细胞外基质(dECM)作为聚对苯二甲酸乙二醇酯(PET)聚合物纤维支架的改性剂来复制细胞生长环境。这使我们能够创建一个dECM涂层的聚酯纤维基支架,PET-dECM,专为肝脏组织工程目的。我们通过涉及胰蛋白酶/EDTA的联合脱细胞过程有效地产生了脱细胞的人脐带源性ECM,TritonX-100和脱氧胆酸钠。dECM涂层在PET材料上的应用是通过几个步骤完成的。如酯水解,EDC/NHS活化交联,和dECM缀合。使用RG细胞培养测定法验证PET-dECM的生物学性能。值得注意的是,dECM涂层显著提高了PET的亲水性和生物相容性,从而帮助细胞粘附,扩散,和功能分化(p<0.05)。进一步发现,在PET-dECM上,HepaRG的肝细胞功能显着增强,这可能归因于dECM促进细胞极性恢复的能力。PET-dECM有望作为有效的肝细胞培养载体,并有可能在肝脏组织工程中找到应用。
    Organ and tissue dysfunction represents a clinically significant condition. By integrating cell biology with materials science, tissue engineering enables the reconstruction and restoration of damaged tissues or organs, offering a noninvasive repair approach. In our study, we replicated the cellular growth environment by utilizing a human umbilical cord-derived decellularized extracellular matrix (dECM) as a modifying agent for the polyethylene terephthalate (PET) polymeric fiber scaffold. This allowed us to create a dECM-coated polyester fiber-based scaffold, PET-dECM, tailored for liver tissue engineering purposes. We effectively produced a decellularized human umbilical cord-derived ECM through a combined decellularization process involving trypsin/EDTA, TritonX-100, and sodium deoxycholate. The application of the dECM coating onto the PET material was accomplished through several steps, such as ester hydrolysis, EDC/NHS-activated crosslinking, and dECM conjugation. The biological performance of the PET-dECM was validated using RG cell culture assays. Notably, the dECM coating significantly improved PET\'s hydrophilicity and biocompatibility, thereby aiding cell adhesion, proliferation, and functional differentiation (p < 0.05). It was further found that the hepatocyte function of HepaRG was significantly enhanced on the PET-dECM, which may be attributed to the dECM\'s ability to facilitate the restoration of cell polarity. The PET-dECM holds promise as an effective hepatocyte culture carrier and could potentially find application in liver tissue engineering.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脱细胞细胞外基质(dECM)水凝胶提供组织特异性微环境,其适应3D体外细胞培养物中的生理细胞表型。然而,它们的形成取决于胶原纤维的形成,限制物理化学性质调节的复杂过程。因此,使用dECM水凝胶实现可重现的结果是一个挑战。这里,我们证明了溶解的肝脏dECM的硫醇化能够通过Michael型加成快速形成共价交联的水凝胶,允许精确控制机械性能和优越的器官型生物活性。各种脱细胞方法的研究表明,用TritonX-100和氢氧化铵处理肝组织导致几乎完全的DNA去除,并显着保留了天然肝蛋白质组。通过1-乙基-3(3-二甲基氨基)丙基碳二亚胺(EDC)/N-羟基琥珀酰亚胺(NHS)偶联L-半胱氨酸产生的硫醇化肝dECM(dECM-SH),其与4臂聚乙二醇(PEG)-马来酰亚胺快速反应以在受控条件下形成光学透明的水凝胶。重要的是,杨氏模量可以通过改变聚合物浓度在1-7kPa之间精确调节,能够在体外细胞培养中密切复制健康和纤维化的肝脏状况。点击dECM-SH水凝胶是细胞相容性的,支持HepG2和HepaRG肝细胞的生长,并促进肝脏特异性功能表型,如增加的代谢活性所证明,与单层培养物和基于胶原蛋白的水凝胶相比,CYP1A2和CYP3A4的活性和排泄功能也是如此。我们的研究结果表明,点击功能化的dECM水凝胶提供了一种高度可控的,用于体外细胞培养应用的常规组织衍生水凝胶的可重复替代。重要声明:由于可变的交联过程,传统的dECM水凝胶在再现性和机械性能控制方面面临挑战。我们介绍了一种基于猪肝脱细胞细胞外基质(dECM)的点击水凝胶,可以绕过这些挑战。在优化肝脏去细胞化以保持ECM后,我们通过迈克尔型加成点击化学整合巯基官能化肝脏dECM与聚乙二醇衍生物,快速启用,室温凝胶化。这提供了对水凝胶的机械和生化特性的增强控制。所得的clickdECM水凝胶模拟肝脏的天然ECM,并表现出更大的机械可调性和易于处理,促进它们在高通量和工业环境中的应用。此外,这些水凝胶显着改善了3D培养中HepaRG衍生肝细胞的功能,提出了用于药物测试应用的肝组织细胞培养模型的进步。
    Decellularized extracellular matrix (dECM) hydrogels provide tissue-specific microenvironments which accommodate physiological cellular phenotypes in 3D in vitro cell cultures. However, their formation hinges on collagen fibrillogenesis, a complex process which limits regulation of physicochemical properties. Hence, achieving reproducible results with dECM hydrogels poses as a challenge. Here, we demonstrate that thiolation of solubilized liver dECM enables rapid formation of covalently crosslinked hydrogels via Michael-type addition, allowing for precise control over mechanical properties and superior organotypic biological activity. Investigation of various decellularization methodologies revealed that treatment of liver tissue with Triton X-100 and ammonium hydroxide resulted in near complete DNA removal with significant retention of the native liver proteome. Chemical functionalization of pepsin-solubilized liver dECMs via 1-ethyl-3(3-dimethylamino)propyl carbodiimide (EDC)/N-hydroxysuccinimide (NHS) coupling of l-Cysteine created thiolated liver dECM (dECM-SH), which rapidly reacted with 4-arm polyethylene glycol (PEG)-maleimide to form optically clear hydrogels under controlled conditions. Importantly, Young\'s moduli could be precisely tuned between 1 - 7 kPa by varying polymer concentrations, enabling close replication of healthy and fibrotic liver conditions in in vitro cell cultures. Click dECM-SH hydrogels were cytocompatible, supported growth of HepG2 and HepaRG liver cells, and promoted liver-specific functional phenotypes as evidenced by increased metabolic activity, as well CYP1A2 and CYP3A4 activity and excretory function when compared to monolayer culture and collagen-based hydrogels. Our findings demonstrate that click-functionalized dECM hydrogels offer a highly controlled, reproducible alternative to conventional tissue-derived hydrogels for in vitro cell culture applications. STATEMENT OF SIGNIFICANCE: Traditional dECM hydrogels face challenges in reproducibility and mechanical property control due to variable crosslinking processes. We introduce a click hydrogel based on porcine liver decellularized extracellular matrix (dECM) that circumnavigates these challenges. After optimizing liver decellularization for ECM retention, we integrated thiol-functionalized liver dECM with polyethylene-glycol derivatives through Michael-type addition click chemistry, enabling rapid, room-temperature gelation. This offers enhanced control over the hydrogel\'s mechanical and biochemical properties. The resultant click dECM hydrogels mimic the liver\'s natural ECM and exhibit greater mechanical tunability and handling ease, facilitating their application in high-throughput and industrial settings. Moreover, these hydrogels significantly improve the function of HepaRG-derived hepatocytes in 3D culture, presenting an advancement for liver tissue cell culture models for drug testing applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:3D打印广泛用于再生医学,有望实现阴道形态恢复和真正的功能重建。间充质干细胞来源的外泌体(MSC-Exos)应用于各种组织的再生。本研究旨在探讨MSCs-Exos在阴道重建中的作用。
    方法:在这项工作中,使用脱细胞细胞外基质(dECM)和明胶甲基丙烯酸酯(GelMA)和丝素蛋白(SF)设计水凝胶。使用桌面立体光刻法构建生物支架。评价了水凝胶的理化性质;进行了一些实验来评估外泌体促进阴道重建的作用,并探讨了这一过程的机制。
    结果:观察到外泌体在水凝胶中在体外和体外的持续释放性质。结果表明,3D支架包裹外泌体对再生阴道组织的血管化和胶囊再生有显著影响。此外,MSCs-Exos强烈促进大鼠阴道重建血管化,可能通过PI3K/AKT信号通路。
    结论:外泌体-水凝胶复合材料的使用改善了阴道组织的上皮再生,血管生成增加,促进平滑肌组织再生。3D打印,封装外泌体的腔内支架可用作阴道重建的无细胞替代治疗策略.
    BACKGROUND: 3D-printing is widely used in regenerative medicine and is expected to achieve vaginal morphological restoration and true functional reconstruction. Mesenchymal stem cells-derived exosomes (MSCs-Exos) were applyed in the regeneration of various tissues. The current study aimed to explore the effctive of MSCs-Exos in vaginal reconstruction.
    METHODS: In this work, hydrogel was designed using decellularized extracellular matrix (dECM) and gelatin methacrylate (GelMA) and silk fibroin (SF). The biological scaffolds were constructed using desktop-stereolithography. The physicochemical properties of the hydrogels were evaluated; Some experiments have been conducted to evaluate exosomes\' effect of promotion vaginal reconstruction and to explore the mechanism in this process.
    RESULTS: It was observed that the sustained release property of exosomes in the hydrogel both in vitro and in vitro.The results revealed that 3D scaffold encapsulating exosomes expressed significant effects on the vascularization and musule regeneration of the regenerative vagina tissue. Also, MSCs-Exos strongly promoted vascularization in the vaginal reconstruction of rats, which may through the PI3K/AKT signaling pathway.
    CONCLUSIONS: The use of exosome-hydrogel composites improved the epithelial regeneration of vaginal tissue, increased angiogenesis, and promoted smooth muscle tissue regeneration. 3D-printed, lumenal scaffold encapsulating exosomes might be used as a cell-free alternative treatment strategy for vaginal reconstruction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号