3D co-culture

3D 共同文化
  • 文章类型: Journal Article
    软骨肉瘤(CHS),也被称为恶性软骨瘤,是仅次于骨肉瘤的第二大常见骨癌。这个肿瘤特别耐化学性和放射性,唯一的治疗选择是手术的边缘。肿瘤免疫微环境显示肿瘤相关巨噬细胞(TAMs)的浸润,有时接近肿瘤质量的50%,主要分化为M2样表型,与不良预后和转移有关。因此,巨噬细胞靶向疗法可能对CHS的管理感兴趣。为了评估这些策略,我们在此建议在甲基纤维素基质中开发两种人类CHS细胞系(JJ012和CH2879)和人类白血病单核细胞系(THP-1)之间的三维(3D)肿瘤共培养模型。这两个模型在巨噬细胞表型方面与体内异种移植模型进行了比较,蛋白聚糖,MMP-9和COX-2表达。最后,米法莫肽,一种作用于TAM的免疫调节剂,在最体外相关的模型上进行了评估:3D共培养CH2879模型。我们的结果表明,现在有可能开发出3D模型,该模型非常准确地模拟体内发现的东西,并有可能评估针对肿瘤细胞成分的治疗方法。
    Chondrosarcoma (CHS), also known as malignant cartilage tumors, is the second most common bone cancer after osteosarcoma. This tumor is particularly chemo- and radioresistant, and the only therapeutic alternative is surgery with wide margins. The tumor immune microenvironment reveals an infiltration of tumor-associated macrophages (TAMs) sometimes approaching 50% of the tumor mass, mainly differentiated into M2-like phenotype and correlated with poor prognosis and metastasis. Thus, macrophage-targeting therapies could have an interest in the management of CHS. To evaluate these strategies, we propose here the development of a three-dimensional (3D) tumoroid co-culture model between two human CHS cell lines (JJ012 and CH2879) and a human leukemia monocytic cell line (THP-1) in a methylcellulose matrix. These two models were compared to the in vivo xenograft models in terms of macrophage phenotypes, proteoglycans, MMP-9, and COX-2 expression. Finally, mifamurtide, an immunomodulator acting on TAMs, was evaluated on the most in vitro relevant model: 3D co-culture CH2879 model. Our results showed that it is now possible to develop 3D models that very accurately mimic what is found in vivo with the possibility of evaluating treatments specific to a tumor cell component.
    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
    细胞外基质(ECM)中生物物理和生化线索的空间组织与相互的细胞-细胞信号传导相一致,对于发育过程中的组织模式至关重要。然而,由于其固有的复杂性,使用现有的体内模型很难阐明单个微环境因素的作用。在这项工作中,我们已经开发了一种微生理系统来在空间上对生物化学进行图案化,生物物理,和沿着上皮化的3D微通道的ECM的基质细胞组成。该技术适用于多种水凝胶组合物并且可缩放至图案化的区域的数量。我们证实,创建不同区域的方法导致了连续的,具有区域界面的退火水凝胶不会阻碍可溶性分子的运输。Further,水凝胶区域之间的界面没有破坏微通道结构,上皮腔形成,或通过无细胞或细胞化微通道的培养基灌注。最后,我们证明了连续上皮管在空间上图案化的肾小管生成发芽进入周围的水凝胶,该水凝胶局限于具有基质细胞群的局部区域。说明细胞间相互作用和信号梯度的空间控制。这种易于使用的系统具有广泛的实用性,可用于模拟与异质水凝胶组合物和/或基质细胞群的三维上皮和内皮组织相互作用,以研究其在发育过程中的机制作用。稳态,或疾病。
    The spatial organization of biophysical and biochemical cues in the extracellular matrix (ECM) in concert with reciprocal cell-cell signaling is vital to tissue patterning during development. However, elucidating the role an individual microenvironmental factor plays using existing in vivo models is difficult due to their inherent complexity. In this work, we have developed a microphysiological system to spatially pattern the biochemical, biophysical, and stromal cell composition of the ECM along an epithelialized 3D microchannel. This technique is adaptable to multiple hydrogel compositions and scalable to the number of zones patterned. We confirmed that the methodology to create distinct zones resulted in a continuous, annealed hydrogel with regional interfaces that did not hinder the transport of soluble molecules. Further, the interface between hydrogel regions did not disrupt microchannel structure, epithelial lumen formation, or media perfusion through an acellular or cellularized microchannel. Finally, we demonstrated spatially patterned tubulogenic sprouting of a continuous epithelial tube into the surrounding hydrogel confined to local regions with stromal cell populations, illustrating spatial control of cell-cell interactions and signaling gradients. This easy-to-use system has wide utility for modeling three-dimensional epithelial and endothelial tissue interactions with heterogeneous hydrogel compositions and/or stromal cell populations to investigate their mechanistic roles during development, homeostasis, or disease.
    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
    至少三分之一的上皮性卵巢癌(OC)患者在诊断时出现腹水,几乎所有患者在复发时都有腹水,尤其是因为OC细胞倾向于在腹腔中扩散,导致腹膜转移。腹水对转移前生态位发育的影响,以及导致间皮癌细胞定植的生物学机制,仍然知之甚少。这里,我们发现,腹水通过影响间皮细胞的形态和破坏它们在细胞周期中的分布来削弱间皮。腹水还通过改变细胞连接的组织而导致间皮完整性的不稳定,但不影响间皮细胞合成N-cadherin和ZO-1。此外,腹水诱导局灶性接触的解体,并导致肌动蛋白细胞骨架重组,这可能取决于Rac1的活性。腹水可以使间皮的ECM蛋白致密化和重组,尤其是纤维蛋白原/纤维蛋白,表明它是OC球体周围纤维蛋白原和纤维蛋白的来源。腹水中的纤维蛋白导致OC球状体与间皮粘附,和腹水促进它们的解聚,然后清除间皮细胞。涉及αV和α5β1整合素。总之,腹水及其纤维蛋白原/纤维蛋白成分会影响间皮的完整性,并促进间皮中OC球体的整合素依赖性植入。
    At least one-third of patients with epithelial ovarian cancer (OC) present ascites at diagnosis and almost all have ascites at recurrence especially because of the propensity of the OC cells to spread in the abdominal cavity leading to peritoneal metastasis. The influence of ascites on the development of pre-metastatic niches, and on the biological mechanisms leading to cancer cell colonization of the mesothelium, remains poorly understood. Here, we show that ascites weakens the mesothelium by affecting the morphology of mesothelial cells and by destabilizing their distribution in the cell cycle. Ascites also causes destabilization of the integrity of mesothelium by modifying the organization of cell junctions, but it does not affect the synthesis of N-cadherin and ZO-1 by mesothelial cells. Moreover, ascites induces disorganization of focal contacts and causes actin cytoskeletal reorganization potentially dependent on the activity of Rac1. Ascites allows the densification and reorganization of ECM proteins of the mesothelium, especially fibrinogen/fibrin, and indicates that it is a source of the fibrinogen and fibrin surrounding OC spheroids. The fibrin in ascites leads to the adhesion of OC spheroids to the mesothelium, and ascites promotes their disaggregation followed by the clearance of mesothelial cells. Both αV and α5β1 integrins are involved. In conclusion ascites and its fibrinogen/fibrin composition affects the integrity of the mesothelium and promotes the integrin-dependent implantation of OC spheroids in the mesothelium.
    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
    在自身免疫性糖尿病发作期间,神经-免疫细胞的相互作用似乎起着重要的作用;然而,目前还没有模型可以在体内或体外跟踪和干扰这些相互作用。二维体外模型提供的信息不足,芯片上的微流体或器官通常具有挑战性。我们在这里提出了我们认为是第一个简单的模型,它提供了共培养胰岛与交感神经和免疫细胞的机会。这个模型是基于我们的冲压设备,可以3D打印(STL文件提供)。由于琼脂糖凝胶中的印记,交感神经元,胰岛,和巨噬细胞可以在允许共聚焦活细胞成像的水平接种在特定位置。在这个协议中,我们提供了在我们的共培养模型中构建和执行活细胞成像实验的说明,包括:1)设计用于在凝胶中进行压印的冲压装置,2)分离交感神经元,胰岛,和巨噬细胞,3)共培养条件,4)如何将其用于活细胞成像,和5)更广泛使用该模型的可能性。总之,我们开发了一种易于使用的共培养模型,可以对交感神经之间的相互作用进行操纵和成像,胰岛,和巨噬细胞。这种新的共培养模型可用于研究神经-免疫细胞-胰岛相互作用,并有助于确定胰腺中神经-免疫相互作用的功能相关性。主要特征•一种新颖的设备,允许交感神经元的3D共培养,胰岛,和免疫细胞•该设备允许捕获小鼠交感神经元之间的实时相互作用,胰岛,和免疫细胞在受控环境中共培养六天后。•本协议使用培养的交感神经元分离自上颈神经节使用以前建立的方法(杰克逊和Tourtellotte,2014)在3D合作文化中。•此方法需要我们自己设计的凝胶冲压设备的3D打印(SciLifeLabFigShareDOI上提供的STL打印文件:10.17044/scilifelab.24073062)。
    During the onset of autoimmune diabetes, nerve-immune cell interactions seem to play an important role; however, there are currently no models to follow and interfere with these interactions over time in vivo or in vitro. Two-dimensional in vitro models provide insufficient information and microfluidics or organs on a chip are usually challenging to work with. We present here what we believe to be the first simple model that provides the opportunity to co-culture pancreatic islets with sympathetic nerves and immune cells. This model is based on our stamping device that can be 3D printed (STL file provided). Due to the imprint in the agarose gel, sympathetic neurons, pancreatic islets, and macrophages can be seeded in specific locations at a level that allows for confocal live-cell imaging. In this protocol, we provide the instructions to construct and perform live cell imaging experiments in our co-culture model, including: 1) design for the stamping device to make the imprint in the gel, 2) isolation of sympathetic neurons, pancreatic islets, and macrophages, 3) co-culture conditions, 4) how this can be used for live cell imaging, and 5) possibilities for wider use of the model. In summary, we developed an easy-to-use co-culture model that allows manipulation and imaging of interactions between sympathetic nerves, pancreatic islets, and macrophages. This new co-culture model is useful to study nerve-immune cell-islet interactions and will help to identify the functional relevance of neuro-immune interactions in the pancreas. Key features • A novel device that allows for 3D co-culture of sympathetic neurons, pancreatic islets, and immune cells • The device allows the capture of live interactions between mouse sympathetic neurons, pancreatic islets, and immune cells in a controlled environment after six days of co-culturing. • This protocol uses cultured sympathetic neurons isolated from the superior cervical ganglia using a previously established method (Jackson and Tourtellotte, 2014) in a 3D co-culture. • This method requires 3D printing of our own designed gel-stamping device (STL print file provided on SciLifeLab FigShare DOI: 10.17044/scilifelab.24073062).
    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
    免疫检查点抑制剂(ICI),例如,靶向程序性细胞死亡蛋白1-配体1(PD-L1)或其受体PD-1,已经显著改善了许多癌症的治疗,但迄今为止在胰腺导管腺癌(PDAC)中失败.巨噬细胞代表PDAC的肿瘤微环境(TME)中最丰富的免疫细胞群之一,能够根据其表型支持或抑制肿瘤进展。为了更好地了解PD-L1/PD-1抑制剂在PDAC中的治疗失败,这项研究在PDAC中动态TME的背景下检查了PD-L1的表达,特别关注巨噬细胞的影响。
    将原发性PDAC组织和相应的肝转移的福尔马林固定和石蜡包埋的组织样品用于免疫组织化学分析。连续切片用检测泛细胞角蛋白的抗体染色,CD68、CD163、CD8和PD-L1。为了研究PD-1/PD-L1轴和巨噬细胞是否有助于PDAC细胞的免疫逃逸,体外建立了富含基质的3D球体共培养模型,使用不同的PDAC细胞系和巨噬细胞亚型以及CD8+T细胞。进行功能和流式细胞术分析以表征细胞群体。
    免疫组织化学分析显示PD-L1主要由基质细胞表达,包括原发性PDAC组织和相应肝转移中的巨噬细胞而非PDAC细胞。值得注意的是,在CD8+T细胞和肿瘤细胞之间的肿瘤病变的侵袭前沿通常发现高局部巨噬细胞丰度和强PD-L1染色.为了研究表达PD-L1的巨噬细胞是否影响PDAC细胞对PD-L1/PD-1抑制剂治疗的反应,我们建立了一个球体模型,该模型包含两种不同的PDAC细胞系和不同比例的体外分化原代M1-或M2-样极化巨噬细胞.根据我们的现场发现,在巨噬细胞而不是PDAC细胞中观察到高PD-L1表达,PDAC细胞的存在进一步增加。PDAC巨噬细胞球状体增强了共培养的CD8+T细胞的效应表型,例如活化标志物的表达和效应分子的释放。特别是与M1样巨噬细胞相比单一培养球体。然而,这与PDAC细胞死亡增强无关.单独使用Durvalumab或Pembrolizumab或与吉西他滨组合的ICI治疗几乎不影响CD8+T细胞的效应表型以及PDAC细胞死亡。因此,尽管在巨噬细胞中有强烈的PD-L1表达,ICI处理没有导致CD8+T细胞的增强的活化和细胞毒性表型。
    总的来说,我们的研究揭示了在ICI存在下PDAC细胞和巨噬细胞相互作用的新见解.
    Immune checkpoint inhibitors (ICI), e.g., targeting programmed cell death protein 1-ligand 1 (PD-L1) or its receptor PD-1, have markedly improved the therapy of many cancers but so far failed in pancreatic ductal adenocarcinoma (PDAC). Macrophages represent one of the most abundant immune cell populations within the tumor microenvironment (TME) of PDAC being able to either support or restrain tumor progression depending on their phenotype. To better understand treatment failure of PD-L1/PD-1 inhibitors in PDAC, this study examined PD-L1 expression in the context of a dynamic TME in PDAC with a particular focus on the impact of macrophages.
    Formalin-fixed and paraffin embedded tissue samples of primary PDAC tissues and corresponding liver metastases were used for immunohistochemical analyses. Serial sections were stained with antibodies detecting Pan-Cytokeratin, CD68, CD163, CD8, and PD-L1.To investigate whether the PD-1/PD-L1 axis and macrophages contribute to immune escape of PDAC cells, a stroma enriched 3D spheroid coculture model was established in vitro, using different PDAC cell lines and macrophages subtypes as well as CD8+ T cells. Functional and flow cytometry analyses were conducted to characterize cell populations.
    Immunohistochemical analyses revealed that PD-L1 is mainly expressed by stroma cells, including macrophages and not PDAC cells in primary PDAC tissues and corresponding liver metastases. Notably, high local abundance of macrophages and strong PD-L1 staining were commonly found at invasion fronts of tumoral lesions between CD8+ T cells and tumor cells. In order to investigate whether PD-L1 expressing macrophages impact the response of PDAC cells to treatment with PD-L1/PD-1 inhibitors, we developed a spheroid model comprising two different PDAC cell lines and different ratios of in vitro differentiated primary M1- or M2-like polarized macrophages. In line with our in situ findings, high PD-L1 expression was observed in macrophages rather than PDAC cells, which was further increased by the presence of PDAC cells. The effector phenotype of co-cultured CD8+ T cells exemplified by expression of activation markers and release of effector molecules was rather enhanced by PDAC macrophage spheroids, particularly with M1-like macrophages compared to mono-culture spheroids. However, this was not associated with enhanced PDAC cell death. ICI treatment with either Durvalumab or Pembrolizumab alone or in combination with Gemcitabine hardly affected the effector phenotype of CD8+ T cells along with PDAC cell death. Thus, despite strong PD-L1 expression in macrophages, ICI treatment did not result in an enhanced activation and cytotoxic phenotype of CD8+ T cells.
    Overall, our study revealed novel insights into the interplay of PDAC cells and macrophages in the presence of ICI.
    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
    长期吸入二氧化硅纳米颗粒(SiNPs)可诱发肺纤维化(PF),然而,潜在的机制仍然难以捉摸。在这里,我们使用Matrigel构建了一个三维(3D)共培养模型,以研究SiNPs暴露后不同细胞之间的相互作用和潜在的调控机制.方法上,我们通过共培养小鼠单核细胞巨噬细胞(RAW264.7)动态观察暴露于SiNPs后细胞形态和迁移的变化,人类非小细胞肺癌细胞(A549),和医学研究委员会细胞株5(MRC-5)在Matrigel中持续24小时。随后,我们检测到核因子κB(NF-κB)的表达,炎症因子和上皮间质转化(EMT)标志物。结果显示SiNPs对细胞产生毒性作用。在3D共同文化状态下,细胞的运动速度和位移增加,细胞迁移能力增强。同时,炎症因子肿瘤坏死因子-α(TNF-α)的表达,白细胞介素-6(IL-6)上调,上皮标志物E-cadherin(E-cad)下调,间充质标志物N-cadherin(N-cad)和成肌纤维细胞标志物α-平滑肌肌动蛋白(α-SMA)表达上调,而SiNPs暴露后NF-κB表达也上调。我们进一步发现细胞在3D共培养状态下更倾向于转分化为肌成纤维细胞。相反,利用NF-κB特异性抑制剂BAY11-7082有效下调TNF-α的表达,IL-6,白细胞介素-1β(IL-1β),N-cad,α-SMA,胶原蛋白-I(COLI),和纤连蛋白(FN),E-cad的表达上调。这些发现表明NF-κB参与调节SiNPs诱导的炎症,EMT,和纤维化在3D共培养状态。
    Long-term inhalation of silica nanoparticles (SiNPs) can induce pulmonary fibrosis (PF), nevertheless, the potential mechanisms remain elusive. Herein, we constructed a three-dimensional (3D) co-culture model by using Matrigel to investigate the interaction among different cells and potential regulatory mechanisms after SiNPs exposure. Methodologically, we dynamically observed the changes in cell morphology and migration after exposure to SiNPs by co-culturing mouse monocytic macrophages (RAW264.7), human non-small cell lung cancer cells (A549), and medical research council cell strain-5 (MRC-5) in Matrigel for 24 h. Subsequently, we detected the expression of nuclear factor kappa B (NF-κB), inflammatory factor and epithelial-mesenchymal transition (EMT) markers. The results showed that SiNPs produced toxic effects on cells. In the 3D co-culture state, the cell\'s movement velocity and displacement increased, and the cell migration ability was enhanced. Meanwhile, the expression of inflammatory factor tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) were upregulated, the epithelial marker E-cadherin (E-cad) was downregulated, the mesenchymal marker N-cadherin (N-cad) and myofibroblast marker alpha-smooth muscle actin (α-SMA) expression were upregulated, while NF-κB expression was also upregulated after SiNPs exposure. We further found that cells were more prone to transdifferentiate into myofibroblasts in the 3D co-culture state. Conversely, utilizing the NF-κB-specific inhibitor BAY 11-7082 effectively downregulated the expression of TNF-α, IL-6, interleukin-1β (IL-1β), N-cad, α-SMA, collagen-I (COL I), and fibronectin (FN), the expression of E-cad was upregulated. These findings suggest that NF-κB is involved in regulating SiNPs-induced inflammatory, EMT, and fibrosis in the 3D co-culture state.
    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
    信号通路分析和对不同刺激的基因反应的研究通常在2D单一培养中进行。然而,在肾小球内,细胞在3D中生长,并参与与不同肾小球细胞类型的直接和旁分泌相互作用。因此,必须谨慎对待2D单文化实验的结果。我们培养了肾小球内皮细胞,在2D/3D单培养和2D/3D共培养中的足细胞和系膜细胞,并分析细胞存活,自组装,基因表达,细胞-细胞相互作用,和使用活/死检测的基因通路,延时分析,批量RNA测序,qPCR,和免疫荧光染色。不需要脚手架,3D肾小球共培养物自组织成球体。与2D共培养物相比,3D共培养物中的足细胞和肾小球内皮细胞特异性标记物和细胞外基质增加。必须明智地选择管家基因,因为许多用于基因表达标准化的基因本身在3D培养条件下受到影响。在3D共培养模型中,足细胞衍生的VEGFA向肾小球内皮细胞的转运证实了细胞间串扰。在3D中增强对肾小球功能重要的基因的表达,与2D相比,对当前使用的2D单一培养的可靠性提出了质疑。因此,肾小球3D共培养可能更适合研究细胞间通讯,体外疾病建模和药物筛选。
    Signaling-pathway analyses and the investigation of gene responses to different stimuli are usually performed in 2D monocultures. However, within the glomerulus, cells grow in 3D and are involved in direct and paracrine interactions with different glomerular cell types. Thus, the results from 2D monoculture experiments must be taken with caution. We cultured glomerular endothelial cells, podocytes and mesangial cells in 2D/3D monocultures and 2D/3D co-cultures and analyzed cell survival, self-assembly, gene expression, cell-cell interaction, and gene pathways using live/dead assay, time-lapse analysis, bulk-RNA sequencing, qPCR, and immunofluorescence staining. Without any need for scaffolds, 3D glomerular co-cultures self-organized into spheroids. Podocyte- and glomerular endothelial cell-specific markers and the extracellular matrix were increased in 3D co-cultures compared to 2D co-cultures. Housekeeping genes must be chosen wisely, as many genes used for the normalization of gene expression were themselves affected in 3D culture conditions. The transport of podocyte-derived VEGFA to glomerular endothelial cells confirmed intercellular crosstalk in the 3D co-culture models. The enhanced expression of genes important for glomerular function in 3D, compared to 2D, questions the reliability of currently used 2D monocultures. Hence, glomerular 3D co-cultures might be more suitable in the study of intercellular communication, disease modelling and drug screening ex vivo.
    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
    细菌性胸膜感染与高死亡率相关。由于生物膜形成,治疗是复杂的。常见的致病病原体是金黄色葡萄球菌(S.金黄色葡萄球菌)。因为它明显是人类特有的,啮齿动物模型不能为研究提供足够的条件。这项研究的目的是使用最近建立的源自人类样本的胸膜的3D器官型共培养模型来检查金黄色葡萄球菌感染对人胸膜间皮细胞的影响。我们的模型感染金黄色葡萄球菌后,在确定的时间点收获样品。紧密连接蛋白的组织学分析和免疫染色(c-Jun,VE-钙黏着蛋白,和ZO-1)进行,表现出与体内脓胸相当的变化。分泌的细胞因子水平的测量(TNF-α,MCP-1和IL-1β)在我们的模型中证明了宿主-病原体的相互作用。同样,间皮细胞在体内水平上产生VEGF。这些发现与重要的,无菌对照模型中的未受损细胞。我们能够建立一个3D器官型体外共培养模型的人胸膜感染金黄色葡萄球菌,导致生物膜的形成,包括宿主-病原体相互作用。这种新型模型可能是一种有用的微环境工具,可用于胸膜脓胸中生物膜的体外研究。
    Bacterial pleural infections are associated with high mortality. Treatment is complicated due to biofilm formation. A common causative pathogen is Staphylococcus aureus (S. aureus). Since it is distinctly human-specific, rodent models do not provide adequate conditions for research. The purpose of this study was to examine the effects of S. aureus infection on human pleural mesothelial cells using a recently established 3D organotypic co-culture model of pleura derived from human specimens. After infection of our model with S. aureus, samples were harvested at defined time points. Histological analysis and immunostaining for tight junction proteins (c-Jun, VE-cadherin, and ZO-1) were performed, demonstrating changes comparable to in vivo empyema. The measurement of secreted cytokine levels (TNF-α, MCP-1, and IL-1β) proved host-pathogen interactions in our model. Similarly, mesothelial cells produced VEGF on in vivo levels. These findings were contrasted by vital, unimpaired cells in a sterile control model. We were able to establish a 3D organotypic in vitro co-culture model of human pleura infected with S. aureus resulting in the formation of biofilm, including host-pathogen interactions. This novel model could be a useful microenvironment tool for in vitro studies on biofilm in pleural empyema.
    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
    胰腺癌(PaCa)具有致密的肿瘤微环境,导致患者预后不良。胰腺星状细胞(PSC)是PaCa微环境中最丰富的细胞,也是胶原蛋白的主要来源。骨膜素,一种体细胞蛋白质,由PSC特异性产生并通过促进细胞外胶原组装来促进PaCa细胞的侵袭性。这里,我们的目的是通过抑制骨膜素来减少细胞外胶原蛋白的组装,从而增加自然杀伤(NK)细胞的细胞毒活性。使用CRISPR-Cas9抑制PSC(称为PSC-P)中的骨膜素表达。将PaCa细胞(BxPC-3)与PSC和PSC-P细胞在3D环境中共培养以形成模拟肿瘤微环境的肿瘤球体。通过Masson三色染色评估球体的细胞外胶原蛋白产生。通过流式细胞术和共聚焦显微镜经由CD107a染色分析NK-92细胞的细胞毒活性。通过使用流式细胞术测量膜联蛋白-V和PI阳性来评估BxPC-3细胞中的细胞死亡。因此,骨膜素抑制减少细胞外胶原,增加NK-92细胞向球体的浸润,并在PaCa细胞中诱导细胞死亡。总之,我们建议骨膜素可能是PaCa的治疗靶点,因此需要使用体内模型进行进一步分析以进行概念验证.
    Pancreatic cancer (PaCa) characteristically has a dense tumor microenvironment, which results in poor patient prognosis. Pancreatic stellate cells (PSCs) are the most abundant cells in the PaCa microenvironment and the principal source of collagen. Periostin, a matricellular protein, is produced specifically by PSCs and promotes the aggressiveness of PaCa cells by facilitating extracellular collagen assembly. Here, we aimed to decrease extracellular collagen assembly by suppressing periostin, thereby increasing the cytotoxic activity of natural killer (NK) cells. Periostin expression was suppressed in PSCs (called PSC-P) using CRISPR-Cas9. PaCa cells (BxPC-3) were co-cultured with PSC and PSC-P cells in a 3D environment to form tumor spheroids mimicking the tumor microenvironment. The extracellular collagen production of spheroids was evaluated by Masson\'s trichrome staining. The cytotoxic activity of NK-92 cells was analyzed by flow cytometry and confocal microscopy via CD107a staining. Cell death in BxPC-3 cells was evaluated by measuring Annexin-V and PI positivity using flow cytometry. As a result, periostin suppression decreased extracellular collagen and increased the infiltration of NK-92 cells into spheroids, and induced cell death in PaCa cells. In conclusion, we suggest that periostin might be a therapeutic target for PaCa and further analysis is warranted using in vivo models for proof-of-concept.
    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:神经内tau蛋白聚集是神经退行性tau蛋白病的主要病理标志。现在普遍承认tau聚集也以细胞非自主方式影响星形胶质细胞。然而,所涉及的机制尚不清楚,部分原因是缺乏反映人类tau病变大脑情况的模型。为了准确模拟tau蛋白病变中神经元-星形胶质细胞的相互作用,需要一个包含人类神经元和星形胶质细胞的模型,神经内tau病理学和模拟大脑的三维结构。
    结果:在这里,我们建立了一种新型的tau病理学的100-200µm厚的3D人类神经元/星形胶质细胞共培养模型,包含微孔形式的hiPSC衍生的神经元和原代人星形胶质细胞的同质群体。使用共聚焦,电子显微镜和活体显微镜,我们通过显示3D共培养中的神经元形成突触前和突触后并在4周内显示自发性钙瞬变来验证该程序。3D共培养中的星形胶质细胞显示出双极和星状形态,其过程广泛,可包裹神经元体,在空间上与轴突和树突对齐,可以在突触周围找到。星形胶质细胞的复杂形态以及3D共培养中与神经元的相互作用反映了人脑中,表明该模型在体外研究生理和病理神经元-星形胶质细胞相互作用的潜力。最后,我们成功地实施了一种方法,在3D共培养中引入种子无关的神经内tau聚集,能够研究早期tau发病机制中的神经元-星形胶质细胞相互作用。
    结论:总而言之,这些数据为这种快速的实用性提供了概念验证,小型化,以及用于细胞类型特定操作的标准化3D模型,例如与神经退行性疾病相关的神经内病理学。
    BACKGROUND: Intraneuronal tau aggregation is the major pathological hallmark of neurodegenerative tauopathies. It is now generally acknowledged that tau aggregation also affects astrocytes in a cell non-autonomous manner. However, mechanisms involved are unclear, partly because of the lack of models that reflect the situation in the human tauopathy brain. To accurately model neuron-astrocyte interaction in tauopathies, there is a need for a model that contains both human neurons and human astrocytes, intraneuronal tau pathology and mimics the three-dimensional architecture of the brain.
    RESULTS: Here we established a novel 100-200 µm thick 3D human neuron/astrocyte co-culture model of tau pathology, comprising homogenous populations of hiPSC-derived neurons and primary human astrocytes in microwell format. Using confocal, electron and live microscopy, we validate the procedures by showing that neurons in the 3D co-culture form pre- and postsynapses and display spontaneous calcium transients within 4 weeks. Astrocytes in the 3D co-culture display bipolar and stellate morphologies with extensive processes that ensheath neuronal somas, spatially align with axons and dendrites and can be found perisynaptically. The complex morphology of astrocytes and the interaction with neurons in the 3D co-culture mirrors that in the human brain, indicating the model\'s potential to study physiological and pathological neuron-astrocyte interaction in vitro. Finally, we successfully implemented a methodology to introduce seed-independent intraneuronal tau aggregation in the 3D co-culture, enabling study of neuron-astrocyte interaction in early tau pathogenesis.
    CONCLUSIONS: Altogether, these data provide proof-of-concept for the utility of this rapid, miniaturized, and standardized 3D model for cell type-specific manipulations, such as the intraneuronal pathology that is associated with neurodegenerative disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号