multiplex imaging

多路成像
  • 文章类型: Journal Article
    成像流式细胞术,它结合了流式细胞术和显微镜的优点,已成为各种生物医学领域(如癌症检测)中细胞分析的强大工具。在这项研究中,我们通过采用空间波分复用技术开发了多重成像流式细胞术(mIFC)。我们的mIFC可以同时获得流中单个细胞的明场和多色荧光图像,由金属卤化物灯激发并由单个检测器测量。分辨率测试镜头多重成像实验的统计分析结果,放大试验镜头,和荧光微球验证了mIFC的操作具有良好的成像通道一致性和微米级区分能力。设计了一种用于多路图像处理的深度学习方法,该方法由三个深度学习网络(U-net,非常深的超分辨率,和视觉几何组19)。证明了分化簇24(CD24)成像通道比明场更敏感,核,或癌抗原125(CA125)成像通道在分类三种类型的卵巢细胞系(IOSE80正常细胞,A2780和OVCAR3癌细胞)。当考虑所有四个成像通道时,通过深度学习分析对这三种类型的细胞进行分类的平均准确率为97.1%。我们的单检测器mIFC有望用于未来成像流式细胞仪的开发以及在各种生物医学领域中通过深度学习进行自动单细胞分析。
    Imaging flow cytometry, which combines the advantages of flow cytometry and microscopy, has emerged as a powerful tool for cell analysis in various biomedical fields such as cancer detection. In this study, we develop multiplex imaging flow cytometry (mIFC) by employing a spatial wavelength division multiplexing technique. Our mIFC can simultaneously obtain brightfield and multi-color fluorescence images of individual cells in flow, which are excited by a metal halide lamp and measured by a single detector. Statistical analysis results of multiplex imaging experiments with resolution test lens, magnification test lens, and fluorescent microspheres validate the operation of the mIFC with good imaging channel consistency and micron-scale differentiation capabilities. A deep learning method is designed for multiplex image processing that consists of three deep learning networks (U-net, very deep super resolution, and visual geometry group 19). It is demonstrated that the cluster of differentiation 24 (CD24) imaging channel is more sensitive than the brightfield, nucleus, or cancer antigen 125 (CA125) imaging channel in classifying the three types of ovarian cell lines (IOSE80 normal cell, A2780, and OVCAR3 cancer cells). An average accuracy rate of 97.1% is achieved for the classification of these three types of cells by deep learning analysis when all four imaging channels are considered. Our single-detector mIFC is promising for the development of future imaging flow cytometers and for the automatic single-cell analysis with deep learning in various biomedical fields.
    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
    肿瘤微环境(TME)对肿瘤生长和免疫治疗效果有显著影响。然而,驱动这些效应的TME内的精确细胞相互作用和空间组织仍然难以捉摸。使用先进的多路成像技术,我们发现调节性T细胞(Tregs)在周围肿瘤基质的淋巴管周围积累。富含免疫调节分子(mregDC)的成熟树突状细胞促进了这种局部积累。促进Tregs的趋化性,建立淋巴周围的Treg-mregDC生态位。在这个利基市场中,mregDC促进Treg激活,这反过来又限制了肿瘤抗原运输到引流的肠系膜淋巴结,从而阻碍抗肿瘤适应性免疫应答的启动。破坏Treg募集到mregDC抑制肿瘤进展。我们的研究为TME的组织以及淋巴样细胞和骨髓细胞之间的局部串扰如何抑制抗肿瘤免疫反应提供了有价值的见解。
    The tumor microenvironment (TME) has a significant impact on tumor growth and immunotherapy efficacies. However, the precise cellular interactions and spatial organizations within the TME that drive these effects remain elusive. Using advanced multiplex imaging techniques, we have discovered that regulatory T cells (Tregs) accumulate around lymphatic vessels in the peripheral tumor stroma. This localized accumulation is facilitated by mature dendritic cells enriched in immunoregulatory molecules (mregDCs), which promote chemotaxis of Tregs, establishing a peri-lymphatic Treg-mregDC niche. Within this niche, mregDCs facilitate Treg activation, which in turn restrains the trafficking of tumor antigens to the draining mesenteric lymph nodes, thereby impeding the initiation of anti-tumor adaptive immune responses. Disrupting Treg recruitment to mregDCs inhibits tumor progression. Our study provides valuable insights into the organization of TME and how local crosstalk between lymphoid and myeloid cells suppresses anti-tumor immune responses.
    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
    多重胶原生物标志物的准确检测对于肿瘤、纤维化等各种危重疾病的诊断和治疗至关重要。尽管量子点(QD)具有诱人的光学特性,创建用于多重生物成像的稳定且特异性的基于量子点的探针在技术上仍然具有挑战性。我们首次报道了基于多色QDs的肽探针的构建,用于结缔组织中多重胶原生物标志物的同时指纹识别。已经开发了由谷胱甘肽(GSH)宿主肽和胶原蛋白靶向客体肽(CTP)组成的双肽系统,产生的CTP-QDs探针在暴露于紫外线照射和弱酸性条件下表现出出色的发光稳定性。通用的双肽系统可以轻松地一锅法合成高质量的多色CTP-QDs探针,在靶向关键胶原生物标志物(包括变性胶原)方面表现出优异的选择性,I型胶原蛋白,II型胶原蛋白,和IV型胶原蛋白。多色CTP-QDs探针在同时对多种结缔组织中的多种胶原蛋白类型进行指纹分析方面表现出了显着的功效。不管他们的地位,是否受到伤害的影响,疾病,或正在经历重塑过程。创新的多色CTP-QDs探针为胶原蛋白家族的多重指纹提供了强大的工具包,在胶原蛋白相关疾病的诊断和治疗中显示出巨大的潜力。
    The accurate detection of multiplex collagen biomarkers is vital for diagnosing and treating various critical diseases such as tumors and fibrosis. Despite the attractive optical properties of quantum dots (QDs), it remains technically challenging to create stable and specific QDs-based probes for multiplex biological imaging. We report for the first time the construction of multi-color QDs-based peptide probes for the simultaneous fingerprinting of multiplex collagen biomarkers in connective tissues. A bipeptide system composed of a glutathione (GSH) host peptide and a collagen-targeting guest peptide (CTP) has been developed, yielding CTP-QDs probes that exhibit exceptional luminescence stability when exposed to ultraviolet irradiation and mildly acidic conditions. The versatile bipeptide system allows for facile one-pot synthesis of high-quality multicolor CTP-QDs probes, exhibiting superior selectivity in targeting critical collagen biomarkers including denatured collagen, type I collagen, type II collagen, and type IV collagen. The multicolor CTP-QDs probes have demonstrated remarkable efficacy in simultaneously fingerprinting multiple collagen types in diverse connective tissues, irrespective of their status, whether affected by injury, diseases, or undergoing remodeling processes. The innovative multicolor CTP-QDs probes offer a robust toolkit for the multiplex fingerprinting of the collagen suprafamily, demonstrating significant potential in the diagnosis and treatment of collagen-related diseases.
    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
    免疫肿瘤学领域的最新进展为癌症患者的管理带来了革命性的变化。已发现肿瘤的免疫谱在预测各种癌症的疾病预后和治疗反应中具有关键价值。多重免疫组织化学和免疫荧光已成为同时检测单个组织切片中多种蛋白质生物标志物的有效工具。从而扩大分子和免疫谱分析的机会,同时保留组织样品。通过确定单个肿瘤细胞在混合细胞群中分布时的表型,通过对肿瘤样本进行高通量多重免疫分型来鉴定临床相关生物标志物,对于指导适当的治疗选择具有巨大潜力.此外,新型多标记物成像方法的出现现在可以为肿瘤微环境提供前所未有的见解,包括各种细胞类型之间的潜在相互作用。然而,在日常研究和临床实践中广泛整合这些技术存在重大挑战。这篇综述从监管和临床试验的角度出发,在结构化的行动框架内解决了挑战和潜在的解决方案。还描述了使用多路复用组织成像平台和相关数字病理学进行免疫分型领域的新进展。特别关注不同癌症亚型的翻译意义。©2024作者由JohnWiley&SonsLtd代表英国和爱尔兰病理学会出版的病理学杂志。
    Recent advances in the field of immuno-oncology have brought transformative changes in the management of cancer patients. The immune profile of tumours has been found to have key value in predicting disease prognosis and treatment response in various cancers. Multiplex immunohistochemistry and immunofluorescence have emerged as potent tools for the simultaneous detection of multiple protein biomarkers in a single tissue section, thereby expanding opportunities for molecular and immune profiling while preserving tissue samples. By establishing the phenotype of individual tumour cells when distributed within a mixed cell population, the identification of clinically relevant biomarkers with high-throughput multiplex immunophenotyping of tumour samples has great potential to guide appropriate treatment choices. Moreover, the emergence of novel multi-marker imaging approaches can now provide unprecedented insights into the tumour microenvironment, including the potential interplay between various cell types. However, there are significant challenges to widespread integration of these technologies in daily research and clinical practice. This review addresses the challenges and potential solutions within a structured framework of action from a regulatory and clinical trial perspective. New developments within the field of immunophenotyping using multiplexed tissue imaging platforms and associated digital pathology are also described, with a specific focus on translational implications across different subtypes of cancer. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of 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

       PDF(Pubmed)

  • 文章类型: Journal Article
    Cell surface proteins, as important components of biological membranes, cover a wide range of important markers of diseases and even cancers. In this regard, precise detection of their expression levels is of crucial importance for both cancer diagnosis and the development of responsive therapeutic strategies. Herein, a size-controlled core-shell Au@ Copper(II) benzene-1,3,5-tricarboxylate (Au@Cu-BTC) nanomaterial was synthesized for specific and simultaneous imaging of multiple protein expression levels on cell membranes. The porous shell of Cu-BTC constructed on Au nanoparticles enabled effective loading of Raman reporter molecules, followed by further modification of the targeting moieties, which equipped the nanoprobe with good specificity and stability. Additionally, given the flexibility of the types of Raman reporter molecules available for loading, the nanoprobes were also demonstrated with good multichannel imaging capabilities. Ultimately, the present strategy of electromagnetic and chemical dual Raman scattering enhancement was successfully applied for the simultaneous detection of varied proteins on cell surfaces with high sensitivity and accuracy. The proposed nanomaterial holds promising applications in biosensing and therapeutic fields, which could not only provide a general strategy for the synthesis of metal-organic framework-based core-shell surface-enhanced Raman scattering nanoprobes but also enable further utilization in multitarget and multichannel cell imaging.
    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
    To maximize tumor excision and minimize collateral damage are the primary goals of cancer surgery. Emerging molecular imaging techniques have made \"image-guided surgery\" developed into \"molecular imaging-guided surgery\", which is termed as \"targeted surgery\" in this review. Consequently, the precision of surgery can be advanced from tissue-scale to molecule-scale, enabling \"targeted surgery\" to be a component of \"targeted therapy\". Evidence from numerous experimental and clinical studies has demonstrated significant benefits of fluorescent imaging in targeted surgery with preoperative molecular diagnostic screening. Fluorescent imaging can help to improve intraoperative staging and enable more radical cytoreduction, detect obscure tumor lesions in special organs, highlight tumor margins, better map lymph node metastases, and identify important normal structures intraoperatively. Though limited tissue penetration of fluorescent imaging and tumor heterogeneity are two major hurdles for current targeted surgery, multimodality imaging and multiplex imaging may provide potential solutions to overcome these issues, respectively. Moreover, though many fluorescent imaging techniques and probes have been investigated, targeted surgery remains at a proof-of-principle stage. The impact of fluorescent imaging on cancer surgery will likely be realized through persistent interdisciplinary amalgamation of research in diverse fields.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号