pancreatic stellate cell

胰腺星状细胞
  • 文章类型: Journal Article
    胰腺癌仍然是最致命的恶性疾病之一。以吉西他滨为基础的化疗仍然是一线全身治疗方法之一,但是化疗耐药发生在大多数患者身上。最近,积累的证据已经证明了肿瘤微环境在促进化疗耐药中的作用。在肿瘤微环境中,胰腺星状细胞(PSC)是主要的细胞成分,和细胞外囊泡(EV)是细胞-细胞通讯的常见介质。在这项研究中,我们发现SP1转录的miR-31-5p不仅靶向胰腺癌细胞中的LATS2,而且还通过EV转移调节PSC中的Hippo通路。因此,PSC合成并分泌酸性和富含半胱氨酸的蛋白质(SPARC),在基质细胞中优先表达,刺激细胞外信号调节激酶(ERK)信号在胰腺癌细胞。因此,由于miR-31-5p调节的内源性Hippo途径和外部SPARC诱导的ERK信号传导,胰腺癌细胞的存活和化学抗性得到改善.在老鼠模型中,miR-31-5p在胰腺癌细胞中的过表达促进了共同注射的异种移植物的化学抗性。在组织微阵列中,miR-31-5p表达较高的胰腺癌患者的总生存期较短.因此,miR-31-5p通过EV调节肿瘤微环境中多种细胞类型的Hippo途径,最终有助于胰腺癌细胞的化学抗性。
    Pancreatic cancer remains one of the most lethal malignant diseases. Gemcitabine-based chemotherapy is still one of the first-line systemic treatments, but chemoresistance occurs in the majority of patients. Recently, accumulated evidence has demonstrated the role of the tumour microenvironment in promoting chemoresistance. In the tumour microenvironment, pancreatic stellate cells (PSCs) are among the main cellular components, and extracellular vesicles (EVs) are common mediators of cell‒cell communication. In this study, we showed that SP1-transcribed miR-31-5p not only targeted LATS2 in pancreatic cancer cells but also regulated the Hippo pathway in PSCs through EV transfer. Consequently, PSCs synthesized and secreted protein acidic and rich in cysteins (SPARC), which was preferentially expressed in stromal cells, stimulating Extracellular Signal regulated kinase (ERK) signalling in pancreatic cancer cells. Therefore, pancreatic cancer cell survival and chemoresistance were improved due to both the intrinsic Hippo pathway regulated by miR-31-5p and external SPARC-induced ERK signalling. In mouse models, miR-31-5p overexpression in pancreatic cancer cells promoted the chemoresistance of coinjected xenografts. In a tissue microarray, pancreatic cancer patients with higher miR-31-5p expression had shorter overall survival. Therefore, miR-31-5p regulates the Hippo pathway in multiple cell types within the tumour microenvironment via EVs, ultimately contributing to the chemoresistance of pancreatic cancer cells.
    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
    建议将胰岛移植作为1型糖尿病(T1D)的治疗方法。尽管它在优化调节葡萄糖水平方面取得了成功,胰岛移植物寿命的限制仍然需要创新的解决方案。移植后的炎性应激和细胞外基质的损失归因于有限的β细胞存活。胰腺星状细胞(PSC),鉴定为胰腺特异性基质细胞,有可能在保护胰岛生存方面发挥关键作用。我们的研究旨在确定在IFN-γ细胞因子混合物诱导的炎症应激下,与人CMβ细胞和人胰岛共培养的PSC的作用。TNF-α和IL-1β。Transwell培养插入物用于评估PSC对β细胞的旁分泌影响,与共同培养一起,使PSC和人类胰岛之间能够直接相互作用。我们发现,将PSC与人CMβ细胞和人尸体胰岛共培养对细胞因子诱导的应激具有挽救作用。在正常血糖和高血糖条件下效果不同。PSC与β细胞线粒体活性的上调和炎症基因表达的抑制有关。在间接和直接共培养方法中都存在挽救作用。此外,我们测试了PSC是否在常规的基于藻酸盐的微胶囊和由藻酸盐-果胶胶原蛋白IV型组成的复合微胶囊中对人类胰岛有挽救作用,层粘连蛋白序列RGD,Nec-1和氨基酸。PSC在两个系统中部分预防细胞因子诱导的应激,但是复合胶囊的有益效果更强。我们的发现显示了PSC对胰岛健康的新作用。胰岛和PSC共培养或共移植可能减轻炎症应激并改善胰岛移植结果。
    Pancreatic islet transplantation is proposed as a cure for type 1 diabetes mellitus (T1D). Despite its success in optimal regulation of glucose levels, limitations in longevity of islet grafts still require innovative solutions. Inflammatory stress post-transplantation and loss of extracellular matrix attribute to the limited β-cell survival. Pancreatic stellate cells (PSCs), identified as pancreatic-specific stromal cells, have the potential to play a crucial role in preserving islet survival. Our study aimed to determine the effects of PSCs co-cultured with human CM β-cells and human islets under inflammatory stress induced by a cytokine cocktail of IFN-γ, TNF-α and IL-1β. Transwell culture inserts were utilized to assess the paracrine impact of PSCs on β-cells, alongside co-cultures enabling direct interaction between PSCs and human islets. We found that co-culturing PSCs with human CM β-cells and human cadaveric islets had rescuing effects on cytokine-induced stress. Effects were different under normoglycemic and hyperglycemic conditions. PSCs were associated with upregulation of β-cell mitochondrial activity and suppression of inflammatory gene expression. The rescuing effects exist both in indirect and direct co-culture methods. Furthermore, we tested whether PSCs have rescuing effects on human islets in conventional alginate-based microcapsules and in composite microcapsules composed of alginate-pectin collagen type IV, laminin sequence RGD, Nec-1, and amino acid. PSCs partially prevented cytokine-induced stress in both systems, but beneficial effects were stronger in composite capsules. Our findings show novel effects of PSCs on islet health. Islets and PSCs coculturing or co-transplantation might mitigate the inflammation stress and improve islet transplantation outcomes.
    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
    胰腺导管腺癌(PDAC)是一种极具侵袭性的癌症,生存率低。成功的治疗策略不应仅限于仅针对癌细胞。但应该采取更全面的方法,考虑其他影响因素。这些包括细胞外基质(ECM)和免疫微环境,两者都是肿瘤微环境的组成部分。本文综述了胰腺星状细胞的作用,分化的癌症相关成纤维细胞和白细胞介素家族,独立或组合,在胰腺上皮内瘤变和PDAC的前体病变进展中。这些元素有助于PDAC中的ECM沉积和免疫抑制。整合白介素和/或基质阻断用于PDAC免疫调节和纤维发生的治疗策略产生了不一致的结果。对纤维化之间复杂的相互作用有更深入的理解,免疫反应可以为更有效的治疗目标铺平道路,通过阐明PDAC进展过程中ECM纤维化的机制和原因。
    Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive form of cancer with a low survival rate. A successful treatment strategy should not be limited to targeting cancer cells alone, but should adopt a more comprehensive approach, taking into account other influential factors. These include the extracellular matrix (ECM) and immune microenvironment, both of which are integral components of the tumor microenvironment. The present review describes the roles of pancreatic stellate cells, differentiated cancer‑associated fibroblasts and the interleukin family, either independently or in combination, in the progression of precursor lesions in pancreatic intraepithelial neoplasia and PDAC. These elements contribute to ECM deposition and immunosuppression in PDAC. Therapeutic strategies that integrate interleukin and/or stromal blockade for PDAC immunomodulation and fibrogenesis have yielded inconsistent results. A deeper comprehension of the intricate interplay between fibrosis, and immune responses could pave the way for more effective treatment targets, by elucidating the mechanisms and causes of ECM fibrosis during PDAC progression.
    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
    胰腺癌的特征在于密集的纤维化基质。纤维化基质阻碍纳米药物的肿瘤内渗透并降低治疗功效。纤维化的特征是细胞外基质(ECM)成分的异常组织,即胶原和纤连蛋白的异常沉积和/或取向。ECM组织异常主要由胰腺星状细胞(PSC)中的病理信号驱动,参与纤维形成的主要细胞类型。然而,涉及ECM异常组织的靶向信号通路是否能改善纳米药物的瘤内渗透尚不清楚.这里,我们表明,靶向转化生长因子-β(TGFβ)/Rho相关激酶(ROCK)1/2信号在PSC正常化ECM组织,并伴随提高纤维化基质的大分子通透性。使用纤维化胰腺癌微环境的三维细胞培养模型,我们发现TGFβ或ROCK1/2的药理学抑制改善了各种大分子的渗透。通过使用同工型特异性药理学抑制剂和siRNA,我们表明,靶向ROCK2,而不是ROCK1,单独足以使ECM组织正常化并改善大分子渗透性。此外,我们发现ROCK2抑制/敲除减弱了与胰腺癌细胞在3D共培养的成纤维细胞中Yes相关蛋白(YAP)的核定位.最后,YAP的药理学抑制或siRNA介导的敲低使ECM组织正常化并改善大分子通透性。我们的结果共同表明,TGFβ/ROCK2/YAP信号轴可能是治疗靶向的,以使ECM组织正常化并改善大分子渗透性,从而增强纳米药物在胰腺癌中的治疗功效。
    Pancreatic cancer is characterized by a densely fibrotic stroma. The fibrotic stroma hinders the intratumoral penetration of nanomedicine and diminishes therapeutic efficacy. Fibrosis is characterized by an abnormal organization of extracellular matrix (ECM) components, namely the abnormal deposition and/or orientation of collagen and fibronectin. Abnormal ECM organization is chiefly driven by pathological signaling in pancreatic stellate cells (PSCs), the main cell type involved in fibrogenesis. However, whether targeting signaling pathways involved in abnormal ECM organization improves the intratumoral penetration of nanomedicines is unknown. Here, we show that targeting transforming growth factor-β (TGFβ)/Rho-associated kinase (ROCK) 1/2 signaling in PSCs normalizes ECM organization and concomitantly improves macromolecular permeability of the fibrotic stroma. Using a 3-dimensional cell culture model of the fibrotic pancreatic cancer microenvironment, we found that pharmacological inhibition of TGFβ or ROCK1/2 improves the permeation of various macromolecules. By using an isoform-specific pharmacological inhibitor and siRNAs, we show that targeting ROCK2, but not ROCK1, alone is sufficient to normalize ECM organization and improve macromolecular permeability. Moreover, we found that ROCK2 inhibition/knockdown attenuates Yes-associated protein (YAP) nuclear localization in fibroblasts co-cultured with pancreatic cancer cells in 3D. Finally, pharmacological inhibition or siRNA-mediated knockdown of YAP normalized ECM organization and improved macromolecular permeability. Our results together suggest that the TGFβ/ROCK2/YAP signaling axis may be therapeutically targeted to normalize ECM organization and improve macromolecular permeability to augment therapeutic efficacy of nanomedicines in pancreatic cancer.
    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
    在纤维炎症领域,慢性胰腺炎(CP)是一种特别具有挑战性的疾病,缺乏专注,批准治疗。吡非尼酮(PFD)的潜力,一种最初用于治疗特发性肺纤维化(IPF)的药物,在解决CP的纤维化方面引发了新的兴趣。这项研究的重点是PFD在减少CP纤维化和免疫反应中的作用,使用caerulein诱导的小鼠模型。该研究扩展到体外研究,研究PFD对胰腺星状细胞(PSC)行为的影响以及巨噬细胞极化为M1和M2类型。采用诸如RNA测序和综合数据分析的先进技术来解码PFD与PSC的分子相互作用。使用定量实时PCR等技术的补充实验,西方印迹,和免疫荧光也被实施。结果显示PFD处理的小鼠胰腺损伤显著减少,表现为腺泡细胞萎缩减少,降低胶原沉积,和减少巨噬细胞的存在。进一步调查显示PFD阻碍PSC迁移的能力,增长,和激活,同时减少了细胞外基质蛋白的产生和分泌。这种作用主要通过干扰TGF-β/Smad等信号通路来实现。Wnt/β-catenin,JAK/STAT此外,PFD通过STAT3途径选择性阻碍M1巨噬细胞极化,不影响M2极化。这些结果强调了PFD在调节PSC活性和M1巨噬细胞极化方面的双重机制,将其定位为CP治疗的有希望的候选人。
    In the realm of fibroinflammatory conditions, chronic pancreatitis (CP) stands out as a particularly challenging ailment, lacking a dedicated, approved treatment. The potential of Pirfenidone (PFD), a drug originally used for treating idiopathic pulmonary fibrosis (IPF), in addressing CP\'s fibrotic aspects has sparked new interest. This investigation focused on the role of PFD in diminishing fibrosis and immune response in CP, using a mouse model induced by caerulein. The research extended to in vitro studies examining the influence of PFD on pancreatic stellate cells\' (PSCs) behavior and the polarization of macrophages into M1 and M2 types. Advanced techniques like RNA sequencing and comprehensive data analyses were employed to decode the molecular interactions of PFD with PSCs. Supplementary experiments using techniques such as quantitative real-time PCR, western blotting, and immunofluorescence were also implemented. Results showed a notable reduction in pancreatic damage in PFD-treated mice, manifested through decreased acinar cell atrophy, lower collagen deposition, and a reduction in macrophage presence. Further investigation revealed PFD\'s capacity to hinder PSCs\' migration, growth, and activation, alongside a reduction in the production and secretion of extracellular matrix proteins. This effect is primarily achieved by interfering with signaling pathways such as TGF-β/Smad, Wnt/β-catenin, and JAK/STAT. Additionally, PFD selectively hampers M1 macrophage polarization through the STAT3 pathway, without impacting M2 polarization. These outcomes highlight PFD\'s dual mechanism in moderating PSC activity and M1 macrophage polarization, positioning it as a promising candidate for CP therapy.
    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
    胰腺星状细胞(PSC)在含有细胞质维生素A(视黄醇)的脂滴丢失后被激活,这是慢性胰腺炎(CP)和胰腺导管腺癌(PDCA)纤维化过程中的关键事件。PSC是产生基质以诱导PDAC癌细胞生长的癌症相关成纤维细胞(CAF)的主要来源,入侵,和转移。作为视黄醇的活性代谢产物,视黄酸(RA)可以通过其核受体复合物(RAR/RXR或RXR/RXR)或转录中介因子调节PSC中的靶基因表达。此外,RA还具有核外和非转录效应。体外研究表明,RA诱导PSC失活,通过多种作用模式减少细胞外基质的产生,如抑制TβRⅡ,PDGFRβ,β-连环蛋白和Wnt产生,下调ERK1/2和JNK磷酸化并抑制活性TGF-β1释放。已证明RA单独或与其他试剂组合对cerulein诱导的小鼠CP模型的体内研究具有有效的抗纤维化作用。临床试验数据表明,将全反式维甲酸(ATRA)重新用作人类胰腺癌的基质靶向剂是安全且可耐受的,提示使用RA治疗人类CP和PDCA的可能性。本文就PSCs中的RA信号通路、RA在PSC介导的纤维化形成中的作用和机制以及靶向PSCs或CAFs的抗纤维化和抗肿瘤作用在体外和体内的研究进展作一综述。强调RA对抗CP和PDAC的潜在疗法。
    Pancreatic stellate cells (PSCs) are activated following loss of cytoplasmic vitamin A (retinol)-containing lipid droplets, which is a key event in the process of fibrogenesis of chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDCA). PSCs are the major source of cancer-associated fibroblasts (CAFs) that produce stroma to induce PDAC cancer cell growth, invasion, and metastasis. As an active metabolite of retinol, retinoic acid (RA) can regulate target gene expression in PSCs through its nuclear receptor complex (RAR/RXR or RXR/RXR) or transcriptional intermediary factor. Additionally, RA also has extranuclear and non-transcriptional effects. In vitro studies have shown that RA induces PSC deactivation which reduces extracellular matrix production through multiple modes of action, such as inhibiting TβRⅡ, PDGFRβ, β-catenin and Wnt production, downregulating ERK1/2 and JNK phosphorylation and suppressing active TGF-β1 release. RA alone or in combination with other reagents have been demonstrated to have an effective anti-fibrotic effect on cerulein-induced mouse CP models in vivo studies. Clinical trial data have shown that repurposing all-trans retinoic acid (ATRA) as a stromal-targeting agent for human pancreatic cancer is safe and tolerable, suggesting the possibility of using RA for the treatment of CP and PDCA in humans. This review focuses on RA signaling pathways in PSCs and the effects and mechanisms of RA in PSC-mediated fibrogenesis as well as the anti-fibrotic and anti-tumor effects of RA targeting PSCs or CAFs in vitro and in vivo, highlighting the potential therapies of RA against CP and PDAC.
    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
    猪胰岛异种移植是一种有希望的治疗严重糖尿病的方法。猪胰岛的长期培养是允许按需提供胰岛的关键挑战。我们旨在确定长期培养成年猪胰岛进行异种移植的最佳温度。我们评估了可能影响在24°C和37°C下成功培养胰岛28天的因素,并发现在37°C下培养有助于胰岛形态的稳定性,胰岛细胞的增殖,以及内分泌功能的恢复,通过参与胰腺发育的基因表达表明,激素产生,和葡萄糖刺激的胰岛素分泌。这些优点可以由胰岛来源的CD146阳性星状细胞提供。使用在37°C下长时间培养的胰岛进行异种移植的功效与过夜培养的胰岛相似。总之,37°C可能是猪胰岛长期培养的合适温度,但是在临床上成功的异种移植需要进一步的修改.
    Porcine islet xenotransplantation represents a promising therapy for severe diabetes mellitus. Long-term culture of porcine islets is a crucial challenge to permit the on-demand provision of islets. We aimed to identify the optimal temperature for the long-term culture of adult porcine islets for xenotransplantation. We evaluated the factors potentially influencing successful 28-day culture of islets at 24°C and 37°C, and found that culture at 37°C contributed to the stability of the morphology of the islets, the proliferation of islet cells, and the recovery of endocrine function, indicated by the expression of genes involved in pancreatic development, hormone production, and glucose-stimulated insulin secretion. These advantages may be provided by islet-derived CD146-positive stellate cells. The efficacy of xenotransplantation using islets cultured for a long time at 37°C was similar to that of overnight-cultured islets. In conclusion, 37°C might be a suitable temperature for the long-term culture of porcine islets, but further modifications will be required for successful xenotransplantation in a clinical setting.
    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
    胰腺星状细胞(PSC)是胰腺中的基质细胞,在胰腺病理中起重要作用。在慢性胰腺炎(CP)和胰腺导管腺癌(PDAC)中,已知PSC被激活以形成促进基质纤维炎症反应的肌成纤维细胞或癌相关成纤维细胞(CAF)。然而,以前对PSC的研究主要基于使用离体扩增的PSC获得的发现,很少有研究使用动物模型解决原位组织驻留PSC的重要性。它们对CP和PDAC中纤维化反应的贡献也鲜为人知。我们对PSC生物学的理解中的这些限制归因于缺乏PSC的特异性分子标记。在这里,我们建立了Meflin(Islr),糖基磷脂酰肌醇锚定膜蛋白,通过使用人胰腺组织样品和Meflin报告小鼠作为小鼠和人的PSC特异性标记。Meflin阳性(Meflin+)细胞含有脂滴并在正常小鼠胰腺中表达常规PSC标记物Desmin,一些细胞对Gli1也呈阳性,Gli1是胰腺组织常驻成纤维细胞的标志物。对Meflin报告小鼠清除胰腺的三维分析显示Meflin+PSC有长而薄的细胞质突起,位于正常胰腺血管的近腔侧。谱系追踪实验表明,Meflin+PSC构成了CP和PDAC中成纤维细胞和CAFs的起源之一,分别。在这些疾病中,Meflin+PSC衍生的成纤维细胞显示出与正常胰腺中的Meflin+PSC不同的形态和分布。此外,我们表明Meflin+PSC的遗传耗竭加速了纤维化,减弱了上皮再生和基质R-spondin3表达,由此暗示Meflin+PSC及其谱系细胞可以支持胰腺损伤和PDAC发育后的组织恢复和Wnt/R-spondin信号传导。一起,这些数据表明Meflin可能是组织驻留PSC的特异性标志物,可用于研究其在健康和疾病中的生物学特性.©2023年英国和爱尔兰病理学会。
    Pancreatic stellate cells (PSCs) are stromal cells in the pancreas that play an important role in pancreatic pathology. In chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC), PSCs are known to get activated to form myofibroblasts or cancer-associated fibroblasts (CAFs) that promote stromal fibroinflammatory reactions. However, previous studies on PSCs were mainly based on the findings obtained using ex vivo expanded PSCs, with few studies that addressed the significance of in situ tissue-resident PSCs using animal models. Their contributions to fibrotic reactions in CP and PDAC are also lesser-known. These limitations in our understanding of PSC biology have been attributed to the lack of specific molecular markers of PSCs. Herein, we established Meflin (Islr), a glycosylphosphatidylinositol-anchored membrane protein, as a PSC-specific marker in both mouse and human by using human pancreatic tissue samples and Meflin reporter mice. Meflin-positive (Meflin+ ) cells contain lipid droplets and express the conventional PSC marker Desmin in normal mouse pancreas, with some cells also positive for Gli1, the marker of pancreatic tissue-resident fibroblasts. Three-dimensional analysis of the cleared pancreas of Meflin reporter mice showed that Meflin+ PSCs have long and thin cytoplasmic protrusions, and are localised on the abluminal side of vessels in the normal pancreas. Lineage tracing experiments revealed that Meflin+ PSCs constitute one of the origins of fibroblasts and CAFs in CP and PDAC, respectively. In these diseases, Meflin+ PSC-derived fibroblasts showed a distinctive morphology and distribution from Meflin+ PSCs in the normal pancreas. Furthermore, we showed that the genetic depletion of Meflin+ PSCs accelerated fibrosis and attenuated epithelial regeneration and stromal R-spondin 3 expression, thereby implying that Meflin+ PSCs and their lineage cells may support tissue recovery and Wnt/R-spondin signalling after pancreatic injury and PDAC development. Together, these data indicate that Meflin may be a marker specific to tissue-resident PSCs and useful for studying their biology in both health and disease. © 2023 The Pathological Society of Great Britain and Ireland.
    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:最近,自然杀伤(NK)细胞成为各种实体瘤的治疗选择。然而,免疫抑制性肿瘤免疫微环境(TIME)可降低胰腺导管腺癌NK细胞的细胞毒性。肿瘤基质内的癌症相关成纤维细胞可以通过与NK细胞的细胞活性有关的失调因子来抑制免疫监视。在这里,研究了活化的胰腺星状细胞(aPSC)在三维(3D)共培养条件下对NK细胞介导的抗癌功效的影响。
    方法:优化了PANC-1肿瘤球体(TS)与aPSC和NK-92细胞在胶原蛋白基质中的3D共培养,以使用微通道芯片鉴定条件培养基中发生的细胞相互作用和差异细胞因子谱。PANC-1TS和aPSC间接共培养,而NK-92细胞被允许使用对流培养基流渗入TS通道。
    结果:与aPSC共培养可促进PANC-1TSs生长并抑制NK-92细胞的抗肿瘤细胞毒作用。在aPSC和NK-92细胞之间观察到细胞活性的相互抑制而不损害迁移能力。此外,发现NK-92细胞的杀伤活性降低与NK细胞中颗粒酶B表达降低有关。
    结论:此处,一种基于PANC-1TS共培养的新型芯片时间模型,aPSC,和NK-92细胞被描述。该模型可用于研究NK细胞失调的详细机制,并探索未来的治疗干预措施以恢复肿瘤微环境中的NK细胞活性。
    BACKGROUND: Recently, natural killer (NK) cells emerged as a treatment option for various solid tumors. However, the immunosuppressive tumor immune microenvironment (TIME) can reduce the cytotoxic ability of NK cells in pancreatic ductal adenocarcinoma. Cancer-associated fibroblasts within the tumor stroma can suppress immune surveillance by dysregulating factors involved in the cellular activity of NK cells. Herein, the effect of activated pancreatic stellate cells (aPSCs) on NK cell-mediated anticancer efficacy under three-dimensional (3D) coculture conditions was investigated.
    METHODS: 3D cocultures of PANC-1 tumor spheroids (TSs) with aPSCs and NK-92 cells in a collagen matrix were optimized to identify the occurring cellular interactions and differential cytokine profiles in conditioned media using microchannel chips. PANC-1 TSs and aPSCs were indirectly cocultured, whereas NK-92 cells were allowed to infiltrate the TS channel using convective medium flow.
    RESULTS: Coculture with aPSCs promoted PANC-1 TSs growth and suppressed the antitumor cytotoxic effects of NK-92 cells. Mutual inhibition of cellular activity without compromising migration ability was observed between aPSCs and NK-92 cells. Moreover, the reduced killing activity of NK-92 cells was found to be related with reduced granzyme B expression in NK cells.
    CONCLUSIONS: Herein, a novel TIME-on-chip model based on the coculture of PANC-1 TSs, aPSCs, and NK-92 cells was described. This model may be useful for studying the detailed mechanisms underlying NK cells dysregulation and for exploring future therapeutic interventions to restore NK cell activity in the tumor microenvironment.
    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
    目的:评价维生素D3类似物卡泊三醇(Cal)对TGF-β1诱导的胰腺星状细胞(PSCs)纤维化的抑制作用及Cal治疗慢性酒精性胰腺炎(ACP)的合理性。
    方法:双标记免疫荧光法用于鉴定健康对照(HC)和ACP患者胰腺中的VDR+PSC。VanGieson染色检查胶原纤维。RT-qPCR和WesternBlot测定VDR的mRNA和蛋白,ACP或体外PSC胰腺中的TGF-β1和COL1A1。ELISA或LC-MS/MS检测血清TGF-β1和COL1A1或25(OH)D3。PSC系(RP-2细胞)用于测定Cal加TGF-β1相对于TGF-β1的蛋白质组改变,并检查VDR基因敲低的作用。
    结果:在用酒精(ALC)加Cal与单独Cal刺激的RP-2细胞中以及在ACP与HC的胰腺中的PSC中检测到VDR的表达增强。增加的VDRPSC与ACP胰腺中COL1A1mRNA水平或胶原沉积区域呈正相关。ACP和ALC处理的RP-2细胞的胰腺中TGF-β1过表达,而ACP与HC相比,血清中25(OH)D3水平显着降低。通过VDR依赖机制,Cal拮抗TGF-β1诱导的RP-2细胞中的16个促纤维化蛋白,包括7个细胞外基质成分,2细胞骨架蛋白,2个纤维化相关因子(RUNX1和TRAF2),TIMP-1,CCN1,整联蛋白α11,一种粘附支架蛋白(TGFB1i1)和一种介导TGF-β1诱导的纤维发生的酶(ENPP1)。
    结论:这项研究表明,Cal给药可能是一种潜在的抗纤维化策略,通过在ACP发展过程中抑制TGF-β1介导的PSC作用。
    OBJECTIVE: To evaluate the inhibitory effect of vitamin D3 analogue calcipotriol (Cal) on the fibrosis of pancreatic stellate cells (PSCs) induced by TGF-β1 and the rationality of Cal use in alcoholic chronic pancreatitis (ACP).
    METHODS: Double-labeling immunofluorescence was used for the identification of VDR+PSCs in the pancreas of healthy controls (HC) and ACP patients. Van Gieson staining for examination of collagen fibers. RT-qPCR and Western Blot for determining the mRNAs and proteins of VDR, TGF-β1 and COL1A1 in the pancreas of ACP or in vitro PSCs. ELISA or LC-MS/MS for detection of serum TGF-β1 and COL1A1 or 25(OH)D3. The PSC line (RP-2 cell) was used for the determination of proteomic alterations in Cal plus TGF-β1 versus TGF-β1 and to examine the effect of VDR gene knockdown.
    RESULTS: Enhanced expression of VDR was detected in RP-2 cells stimulated with alcohol (ALC) plus Cal versus Cal alone and in PSCs in the pancreas of ACP versus HC. The increased VDR+PSCs were positively correlated with the levels of COL1A1 mRNAs or areas of collagen deposition in the pancreas of ACP. TGF-β1 was overexpressed in the pancreas of ACP and ALC-treated RP-2 cells while 25(OH)D3 level in serum was significantly decreased in ACP versus HC. Through a VDR-dependent mechanism, Cal antagonized 16 profibrotic proteins in TGF-β1-induced RP-2 cells that included 7 extracellular matrix components, 2 cytoskeletal proteins, 2 fibrosis-associated factors (RUNX1 and TRAF2), TIMP-1, CCN1, integrin α11, an adhesion scaffold protein (TGFB1i1) and an enzyme mediating TGF-β1-induced fibrogenesis (ENPP1).
    CONCLUSIONS: This study suggests that Cal administration may be a potential antifibrotic strategy via inhibiting TGF-β1-mediated PSC action during the development of ACP.
    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