spatial biology

空间生物学
  • 文章类型: Journal Article
    在组织工程中用于开发生物等效物的所有材料中,胶原蛋白由于其卓越的生物相容性和可生物降解性而被证明是最有前途的,从而成为脚手架生产中应用最广泛的材料之一。然而,目前胶原支架内细胞的成像技术有几个局限性,这导致迫切需要新的可视化方法。在这项工作中,我们获得了一组胶原支架,并选择了对比剂,以便通过X射线计算机显微断层扫描(micro-CT)以非破坏性方式研究细胞生长的孔和模式。经过多种对比剂的比较,在蒸馏水中的3%磷钨酸水溶液被确定为最有效的介质,需要24小时的孵化。胶原纤维之间强度值的差异,毛孔,和大量细胞允许进一步分析所需的准确分割。此外,提出的协议允许多孔胶原支架在水性条件下的可视化,这对于样品天然结构的多模态研究至关重要。
    Among all of the materials used in tissue engineering in order to develop bioequivalents, collagen shows to be the most promising due to its superb biocompatibility and biodegradability, thus becoming one of the most widely used materials for scaffold production. However, current imaging techniques of the cells within collagen scaffolds have several limitations, which lead to an urgent need for novel methods of visualization. In this work, we have obtained groups of collagen scaffolds and selected the contrasting agents in order to study pores and patterns of cell growth in a non-disruptive manner via X-ray computed microtomography (micro-CT). After the comparison of multiple contrast agents, a 3% aqueous phosphotungstic acid solution in distilled water was identified as the most effective amongst the media, requiring 24 h of incubation. The differences in intensity values between collagen fibers, pores, and masses of cells allow for the accurate segmentation needed for further analysis. Moreover, the presented protocol allows visualization of porous collagen scaffolds under aqueous conditions, which is crucial for the multimodal study of the native structure of samples.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    空间蛋白质组学能够以单细胞分辨率详细分析组织。然而,创建可靠的分割掩码并将准确的细胞表型分配给离散的细胞表型可能具有挑战性。我们介绍IMmuneCite,用于全面图像预处理和单细胞数据集创建的计算框架,当使用空间蛋白质组学平台时,专注于定义复杂的免疫景观。我们证明IMmuneCite有助于使用来自人类肝脏样品的数据鉴定32个离散的免疫细胞表型,同时大大减少了由不同细胞谱系的标记物的共定位引起的非生物细胞簇。通过将IMmuneCite应用于来自鼠肝组织的数据,我们建立了其多功能性和适应任何抗体组和不同物种的能力。这种方法能够对每个免疫区室中的不同功能状态进行深入表征,揭示临床肝移植和小鼠肝细胞癌中免疫微环境的关键特征。总之,我们证明了IMmuneCite是一个用户友好的,集成的计算平台,有助于研究跨物种的免疫微环境,在确保创造一个专注于免疫的人的同时,空间分辨的单细胞蛋白质组数据集以提供高保真度,生物学相关分析。
    Spatial proteomics enable detailed analysis of tissue at single cell resolution. However, creating reliable segmentation masks and assigning accurate cell phenotypes to discrete cellular phenotypes can be challenging. We introduce IMmuneCite, a computational framework for comprehensive image pre-processing and single-cell dataset creation, focused on defining complex immune landscapes when using spatial proteomics platforms. We demonstrate that IMmuneCite facilitates the identification of 32 discrete immune cell phenotypes using data from human liver samples while substantially reducing nonbiological cell clusters arising from co-localization of markers for different cell lineages. We established its versatility and ability to accommodate any antibody panel and different species by applying IMmuneCite to data from murine liver tissue. This approach enabled deep characterization of different functional states in each immune compartment, uncovering key features of the immune microenvironment in clinical liver transplantation and murine hepatocellular carcinoma. In conclusion, we demonstrated that IMmuneCite is a user-friendly, integrated computational platform that facilitates investigation of the immune microenvironment across species, while ensuring the creation of an immune focused, spatially resolved single-cell proteomic dataset to provide high fidelity, biologically relevant analyses.
    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:糖尿病肾病(DKD)是美国乃至全世界慢性和终末期肾病的主要病因。动物模型教会了我们很多关于DKD机制的知识,但是,由于我们对人类DKD的分子理解滞后,因此将这些知识转化为人类疾病的治疗方法。
    方法:使用我们的空间组织蛋白质组学(STEP)管道(包括经过筛选的人类肾脏组织,多重免疫荧光和强大的分析工具),我们成像并分析了来自糖尿病和健康肾脏的23个组织切片中21种蛋白质的表达(n=5),与DKDIIA相比,IIA-B和IIB(各n=2)和DKDIII(n=1)。
    结果:这些分析显示存在11个细胞簇(肾脏区室/细胞类型):足细胞,肾小球内皮细胞,近端小管,远端肾单位,肾小管周围毛细血管,血管(内皮细胞和血管平滑肌细胞),巨噬细胞,骨髓细胞,其他CD45+炎症细胞,基底膜和间质。DKD进展与炎症细胞和胶原IV沉积的共定位增加相关,伴随着每个肾单位段的天然蛋白质的损失。细胞类型频率和邻域分析强调了DKD中炎性细胞的显着增加及其与肾小管和αSMA(α-平滑肌肌动蛋白阳性)细胞的邻接。最后,DKD进展在单个组织切片中显示出明显的区域变异性,以及每个DKD类别内的个体差异。
    结论:使用STEP管道,我们发现了蛋白质表达的改变,细胞表型组成和微环境结构与DKD进展,展示了这一管道揭示人类DKD病理生理学的力量。
    OBJECTIVE: Diabetic kidney disease (DKD) is the leading cause of chronic and end-stage kidney disease in the USA and worldwide. Animal models have taught us much about DKD mechanisms, but translation of this knowledge into treatments for human disease has been slowed by the lag in our molecular understanding of human DKD.
    METHODS: Using our Spatial TissuE Proteomics (STEP) pipeline (comprising curated human kidney tissues, multiplexed immunofluorescence and powerful analysis tools), we imaged and analysed the expression of 21 proteins in 23 tissue sections from individuals with diabetes and healthy kidneys (n=5), compared to those with DKDIIA, IIA-B and IIB (n=2 each) and DKDIII (n=1).
    RESULTS: These analyses revealed the existence of 11 cellular clusters (kidney compartments/cell types): podocytes, glomerular endothelial cells, proximal tubules, distal nephron, peritubular capillaries, blood vessels (endothelial cells and vascular smooth muscle cells), macrophages, myeloid cells, other CD45+ inflammatory cells, basement membrane and the interstitium. DKD progression was associated with co-localised increases in inflammatory cells and collagen IV deposition, with concomitant loss of native proteins of each nephron segment. Cell-type frequency and neighbourhood analyses highlighted a significant increase in inflammatory cells and their adjacency to tubular and αSMA+ (α-smooth muscle actin-positive) cells in DKD. Finally, DKD progression showed marked regional variability within single tissue sections, as well as inter-individual variability within each DKD class.
    CONCLUSIONS: Using the STEP pipeline, we found alterations in protein expression, cellular phenotypic composition and microenvironment structure with DKD progression, demonstrating the power of this pipeline to reveal the pathophysiology of human DKD.
    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
    空间转录组学(ST)是了解组织生物学和疾病机制的强大工具。然而,由于所需的高级数据分析和编程技能,其潜力往往未得到充分利用。为了解决这个问题,我们提出了空间GE,简化ST数据分析的Web应用程序。应用程序spatialGE提供了一个用户友好的界面,可以通过各种分析管道指导没有编程专业知识的用户,包括质量控制,归一化,域检测,表型,和多种空间分析。它还可以在样品之间进行比较分析,并支持各种ST技术。我们通过将其应用于研究黑色素瘤脑转移和默克尔细胞癌的肿瘤微环境,证明了spatialGE的实用性。我们的结果突出了spatialGE识别空间基因表达模式和富集的能力,为更广泛的科学界提供对肿瘤微环境及其在民主化ST数据分析中的实用性的有价值的见解。
    Spatial transcriptomics (ST) is a powerful tool for understanding tissue biology and disease mechanisms. However, its potential is often underutilized due to the advanced data analysis and programming skills required. To address this, we present spatialGE, a web application that simplifies the analysis of ST data. The application spatialGE provides a user-friendly interface that guides users without programming expertise through various analysis pipelines, including quality control, normalization, domain detection, phenotyping, and multiple spatial analyses. It also enables comparative analysis among samples and supports various ST technologies. We demonstrate the utility of spatialGE through its application in studying the tumor microenvironment of melanoma brain metastasis and Merkel cell carcinoma. Our results highlight the ability of spatialGE to identify spatial gene expression patterns and enrichments, providing valuable insights into the tumor microenvironment and its utility in democratizing ST data analysis for the wider scientific community.
    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
    识别细胞类型和状态仍然是一个耗时的过程,空间生物学容易出错的挑战。随着深度学习的使用越来越多,由于细胞水平的可变性,很难一概而论,邻里,以及健康和疾病的利基。为了解决这个问题,我们开发了TACIT,一种无监督的细胞注释算法,使用预定义的签名,在没有训练数据的情况下运行。TACIT使用无偏阈值来区分阳性细胞和背景,专注于相关标记,以在多体分析中识别模糊的细胞。使用来自三个生态位(大脑,肠,压盖),TACIT在准确性和可扩展性方面优于现有的无监督方法。将TACIT识别的细胞类型与新型Shiny应用程序整合在两种炎症性腺体疾病中揭示了新的表型。最后,结合空间转录组学和蛋白质组学,我们在感兴趣的区域发现了不足和过多的免疫细胞类型和状态,表明多模态对于将空间生物学转化为临床应用至关重要。
    Identifying cell types and states remains a time-consuming, error-prone challenge for spatial biology. While deep learning is increasingly used, it is difficult to generalize due to variability at the level of cells, neighborhoods, and niches in health and disease. To address this, we developed TACIT, an unsupervised algorithm for cell annotation using predefined signatures that operates without training data. TACIT uses unbiased thresholding to distinguish positive cells from background, focusing on relevant markers to identify ambiguous cells in multiomic assays. Using five datasets (5,000,000-cells; 51-cell types) from three niches (brain, intestine, gland), TACIT outperformed existing unsupervised methods in accuracy and scalability. Integrating TACIT-identified cell types with a novel Shiny app revealed new phenotypes in two inflammatory gland diseases. Finally, using combined spatial transcriptomics and proteomics, we discovered under- and overrepresented immune cell types and states in regions of interest, suggesting multimodality is essential for translating spatial biology to clinical applications.
    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
    头颈部鳞状细胞癌(HNSCC)是全球最常见的肿瘤之一,人乳头瘤病毒(HPV)感染有助于癌症的发展。传统疗法只能达到有限的效率,尤其是复发或转移性HNSCC。由于免疫景观决定性地影响患者的生存和治疗效果,这项研究全面调查了免疫肿瘤微环境(TME)及其与患者预后的关系,特别关注几种树突状细胞(DC)和T淋巴细胞亚群。因此,56例HNSCC患者的福尔马林固定石蜡包埋肿瘤样本,接受过切除和辅助放疗的人,通过多重免疫组织化学分析DCs的详细表型特征和空间分布,CD8+T细胞,和不同肿瘤区室的T辅助细胞亚群。免疫细胞密度和比例与整个HNSCC队列和不同的HPV或缺氧相关亚组的临床特征相关。浆细胞样DC和T淋巴细胞高度浸润肿瘤基质。在T辅助细胞和CD8+T细胞中,表达程序性细胞死亡蛋白-1(PD-1+)和/或淋巴细胞活化基因-3(LAG-3+)的基质调节性T细胞和上皮内耗尽的CD8+T细胞是主要的表型,表明免疫抑制性TME。HPV相关肿瘤显示I型和II型常规DC(cDC1,cDC2)以及几种CD8T细胞表型(包括耗尽,激活,和增殖T细胞。相反,具有缺氧相关基因特征的肿瘤对这些免疫细胞的浸润减少.通过多元Cox回归,确定了免疫相关的预后因素.由DC和T淋巴细胞的高浸润结合HPV阳性或低缺氧定义的患者簇显示出显著延长的存活。因此,cDC1和CD8+T细胞是局部和远处复发的独立预后因素。这些结果可能有助于实施预测HNSCC患者生存的免疫细胞浸润评分,这种患者分层可能会改善未来个性化放化疗(免疫)治疗的设计。
    Head and neck squamous cell carcinoma (HNSCC) is one of the most common tumor entities worldwide, with human papillomavirus (HPV) infection contributing to cancer development. Conventional therapies achieve only limited efficiency, especially in recurrent or metastatic HNSCC. As the immune landscape decisively impacts the survival of patients and treatment efficacy, this study comprehensively investigated the immunological tumor microenvironment (TME) and its association with patient outcome, with special focus on several dendritic cell (DC) and T lymphocyte subpopulations. Therefore, formalin-fixed paraffin-embedded tumor samples of 56 HNSCC patients, who have undergone resection and adjuvant radiotherapy, were analyzed by multiplex immunohistochemistry focusing on the detailed phenotypic characterization and spatial distribution of DCs, CD8+ T cells, and T-helper cell subsets in different tumor compartments. Immune cell densities and proportions were correlated with clinical characteristics of the whole HNSCC cohort and different HPV- or hypoxia-associated subcohorts. Tumor stroma was highly infiltrated by plasmacytoid DCs and T lymphocytes. Among the T-helper cells and CD8+ T cells, stromal regulatory T cells and intraepithelial exhausted CD8+ T cells expressing programmed cell death protein-1 (PD-1+) and/or lymphocyte-activation gene-3 (LAG-3+) were the predominant phenotypes, indicating an immunosuppressive TME. HPV-associated tumors showed significantly higher infiltration of type I and type II conventional DCs (cDC1, cDC2) as well as several CD8+ T cell phenotypes including exhausted, activated, and proliferating T cells. On the contrary, tumors with hypoxia-associated gene signatures exhibited reduced infiltration for these immune cells. By multivariate Cox regression, immune-related prognostic factors were identified. Patient clusters defined by high infiltration of DCs and T lymphocytes combined with HPV positivity or low hypoxia showed significantly prolonged survival. Thereby, cDC1 and CD8+ T cells emerged as independent prognostic factors for local and distant recurrence. These results might contribute to the implementation of an immune cell infiltration score predicting HNSCC patients\' survival and such patient stratification might improve the design of future individualized radiochemo-(immuno)therapies.
    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
    识别细胞类型和状态对于空间生物学来说仍然是耗时且容易出错的挑战。随着深度学习的使用越来越多,由于细胞水平的可变性,很难一概而论,邻里,以及健康和疾病的利基。为了解决这个问题,我们开发了TACIT,一种无监督的细胞注释算法,使用预定义的签名,在没有训练数据的情况下运行,使用无偏阈值将阳性细胞与背景区分开来,专注于相关标记,以在多体分析中识别模糊的细胞。使用来自三个生态位(大脑,肠,压盖),TACIT在准确性和可扩展性方面优于现有的无监督方法。TACIT鉴定的细胞与新型Shiny应用程序的整合揭示了两种炎症性腺体疾病的新表型。最后,结合空间转录组学和蛋白质组学,我们在感兴趣的区域发现了不足和过多的免疫细胞类型和状态,表明多模态对于将空间生物学转化为临床应用至关重要。
    Identifying cell types and states remains a time-consuming and error-prone challenge for spatial biology. While deep learning is increasingly used, it is difficult to generalize due to variability at the level of cells, neighborhoods, and niches in health and disease. To address this, we developed TACIT, an unsupervised algorithm for cell annotation using predefined signatures that operates without training data, using unbiased thresholding to distinguish positive cells from background, focusing on relevant markers to identify ambiguous cells in multiomic assays. Using five datasets (5,000,000-cells; 51-cell types) from three niches (brain, intestine, gland), TACIT outperformed existing unsupervised methods in accuracy and scalability. Integration of TACIT-identified cell with a novel Shiny app revealed new phenotypes in two inflammatory gland diseases. Finally, using combined spatial transcriptomics and proteomics, we discover under- and overrepresented immune cell types and states in regions of interest, suggesting multimodality is essential for translating spatial biology to clinical applications.
    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
    肿瘤细胞和宿主浸润细胞之间的空间关系在基础科学和临床研究中越来越重要。在这项研究中,我们已经测试了在多重染色系统中使用标准免疫组织化学(IHC)方法的可行性,该方法使用了一组新开发的过氧化物酶和碱性磷酸酶的显色底物。使用这种方法,我们开发了一套色原,其特征是(1)提供精细的细胞细节,(2)非重叠光谱轮廓,(3)色原之间没有相互作用,(4)储存时的稳定性,和(5)与当前标准免疫组织化学实践的相容性。在明场照明下显微观察时,色原产生以下颜色:红色,黑色,蓝色,黄色,棕色,和绿色。通过选择兼容的颜色组合,我们已经证明了四色多重染色的可行性。根据所执行的特定分析类型,可视化分析,没有计算机辅助图像分析的帮助,足以区分多达四种不同的标记。
    Spatial relations between tumor cells and host-infiltrating cells are increasingly important in both basic science and clinical research. In this study, we have tested the feasibility of using standard methods of immunohistochemistry (IHC) in a multiplex staining system using a newly developed set of chromogenic substrates for the peroxidase and alkaline phosphatase enzymes. Using this approach, we have developed a set of chromogens characterized by (1) providing fine cellular detail, (2) non-overlapping spectral profiles, (3) an absence of interactions between chromogens, (4) stability when stored, and (5) compatibility with current standard immunohistochemistry practices. When viewed microscopically under brightfield illumination, the chromogens yielded the following colors: red, black, blue, yellow, brown, and green. By selecting compatible color combinations, we have shown feasibility for four-color multiplex staining. Depending on the particular type of analysis being performed, visual analysis, without the aid of computer-assisted image analysis, was sufficient to differentiate up to four different markers.
    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)是发病率增加的唯一癌症之一,通常与肿瘤内和肿瘤周围的瘢痕形成有关。称为结缔组织增生。这种瘢痕形成在细胞外基质(ECM)结构中是高度异质的,并且在尚未完全理解的肿瘤生物学和临床结果两者中起复杂的作用。使用苏木精和伊红(H&E),现有临床工作流程中使用的常规组织学染色,我们量化了85例患者样本中的ECM结构,以评估促纤维增生性结构与临床结局(如生存时间和疾病复发)之间的关系.通过利用无监督机器学习(ML)来总结147个局部的潜在空间(例如光纤长度,坚固性)和全局(例如纤维分支,孔隙度)基于H&E的特征,我们发现了一系列与生存率和复发率差异相关的组织学结构.Further,我们通过indexing(CODEX)参考图集将H&E架构映射到CO-Detection,揭示与结果阳性和阳性相关的基于细胞和蛋白质的局部生态位肿瘤微环境中的结果阴性瘢痕形成。总的来说,我们的研究利用标准H&E染色来揭示促纤维化组织和PDAC结局之间的临床相关关联,提供了一个可翻译的管道来支持预后决策和空间生物因素的蓝图,用于通过组织工程方法进行建模。
    Pancreatic ductal adenocarcinoma (PDAC) represents one of the only cancers with an increasing incidence rate and is often associated with intra- and peri-tumoral scarring, referred to as desmoplasia. This scarring is highly heterogeneous in extracellular matrix (ECM) architecture and plays complex roles in both tumor biology and clinical outcomes that are not yet fully understood. Using hematoxylin and eosin (H&E), a routine histological stain utilized in existing clinical workflows, we quantified ECM architecture in 85 patient samples to assess relationships between desmoplastic architecture and clinical outcomes such as survival time and disease recurrence. By utilizing unsupervised machine learning to summarize a latent space across 147 local (e.g., fiber length, solidity) and global (e.g., fiber branching, porosity) H&E-based features, we identified a continuum of histological architectures that were associated with differences in both survival and recurrence. Furthermore, we mapped H&E architectures to a CO-Detection by indEXing (CODEX) reference atlas, revealing localized cell- and protein-based niches associated with outcome-positive versus outcome-negative scarring in the tumor microenvironment. Overall, our study utilizes standard H&E staining to uncover clinically relevant associations between desmoplastic organization and PDAC outcomes, offering a translatable pipeline to support prognostic decision-making and a blueprint of spatial-biological factors for modeling by tissue engineering methods.
    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