Pub Date : 2024-05-20Epub Date: 2024-04-26DOI: 10.1016/j.crmeth.2024.100760
Alison B Ross, Darvesh Gorhe, Jenny Kim Kim, Stefanie Hodapp, Lela DeVine, Karina M Chan, Iok In Christine Chio, Marko Jovanovic, Marina Ayres Pereira
The role of protein turnover in pancreatic ductal adenocarcinoma (PDA) metastasis has not been previously investigated. We introduce dynamic stable-isotope labeling of organoids (dSILO): a dynamic SILAC derivative that combines a pulse of isotopically labeled amino acids with isobaric tandem mass-tag (TMT) labeling to measure proteome-wide protein turnover rates in organoids. We applied it to a PDA model and discovered that metastatic organoids exhibit an accelerated global proteome turnover compared to primary tumor organoids. Globally, most turnover changes are not reflected at the level of protein abundance. Interestingly, the group of proteins that show the highest turnover increase in metastatic PDA compared to tumor is involved in mitochondrial respiration. This indicates that metastatic PDA may adopt alternative respiratory chain functionality that is controlled by the rate at which proteins are turned over. Collectively, our analysis of proteome turnover in PDA organoids offers insights into the mechanisms underlying PDA metastasis.
{"title":"Systematic analysis of proteome turnover in an organoid model of pancreatic cancer by dSILO.","authors":"Alison B Ross, Darvesh Gorhe, Jenny Kim Kim, Stefanie Hodapp, Lela DeVine, Karina M Chan, Iok In Christine Chio, Marko Jovanovic, Marina Ayres Pereira","doi":"10.1016/j.crmeth.2024.100760","DOIUrl":"10.1016/j.crmeth.2024.100760","url":null,"abstract":"<p><p>The role of protein turnover in pancreatic ductal adenocarcinoma (PDA) metastasis has not been previously investigated. We introduce dynamic stable-isotope labeling of organoids (dSILO): a dynamic SILAC derivative that combines a pulse of isotopically labeled amino acids with isobaric tandem mass-tag (TMT) labeling to measure proteome-wide protein turnover rates in organoids. We applied it to a PDA model and discovered that metastatic organoids exhibit an accelerated global proteome turnover compared to primary tumor organoids. Globally, most turnover changes are not reflected at the level of protein abundance. Interestingly, the group of proteins that show the highest turnover increase in metastatic PDA compared to tumor is involved in mitochondrial respiration. This indicates that metastatic PDA may adopt alternative respiratory chain functionality that is controlled by the rate at which proteins are turned over. Collectively, our analysis of proteome turnover in PDA organoids offers insights into the mechanisms underlying PDA metastasis.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11133751/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140874809","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-20Epub Date: 2024-05-06DOI: 10.1016/j.crmeth.2024.100764
Shelby L Hooe, Christopher M Green, Kimihiro Susumu, Michael H Stewart, Joyce C Breger, Igor L Medintz
Co-assembling enzymes with nanoparticles (NPs) into nanoclusters allows them to access channeling, a highly efficient form of multienzyme catalysis. Using pyruvate kinase (PykA) and lactate dehydrogenase (LDH) to convert phosphoenolpyruvic acid to lactic acid with semiconductor quantum dots (QDs) confirms how enzyme cluster formation dictates the rate of coupled catalytic flux (kflux) across a series of differentially sized/shaped QDs and 2D nanoplatelets (NPLs). Enzyme kinetics and coupled flux were used to demonstrate that by mixing different NP systems into clusters, a >10× improvement in kflux is observed relative to free enzymes, which is also ≥2× greater than enhancement on individual NPs. Cluster formation was characterized with gel electrophoresis and transmission electron microscopy (TEM) imaging. The generalizability of this mixed-NP approach to improving flux is confirmed by application to a seven-enzyme system. This represents a powerful approach for accessing channeling with almost any choice of enzymes constituting a multienzyme cascade.
{"title":"Optimizing the conversion of phosphoenolpyruvate to lactate by enzymatic channeling with mixed nanoparticle display.","authors":"Shelby L Hooe, Christopher M Green, Kimihiro Susumu, Michael H Stewart, Joyce C Breger, Igor L Medintz","doi":"10.1016/j.crmeth.2024.100764","DOIUrl":"10.1016/j.crmeth.2024.100764","url":null,"abstract":"<p><p>Co-assembling enzymes with nanoparticles (NPs) into nanoclusters allows them to access channeling, a highly efficient form of multienzyme catalysis. Using pyruvate kinase (PykA) and lactate dehydrogenase (LDH) to convert phosphoenolpyruvic acid to lactic acid with semiconductor quantum dots (QDs) confirms how enzyme cluster formation dictates the rate of coupled catalytic flux (k<sub>flux</sub>) across a series of differentially sized/shaped QDs and 2D nanoplatelets (NPLs). Enzyme kinetics and coupled flux were used to demonstrate that by mixing different NP systems into clusters, a >10× improvement in k<sub>flux</sub> is observed relative to free enzymes, which is also ≥2× greater than enhancement on individual NPs. Cluster formation was characterized with gel electrophoresis and transmission electron microscopy (TEM) imaging. The generalizability of this mixed-NP approach to improving flux is confirmed by application to a seven-enzyme system. This represents a powerful approach for accessing channeling with almost any choice of enzymes constituting a multienzyme cascade.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11133815/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140877466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-20Epub Date: 2024-05-13DOI: 10.1016/j.crmeth.2024.100776
Sarah Cooper, Sofia Obolenski, Andrew J Waters, Andrew R Bassett, Matthew A Coelho
Continual advancements in genomics have led to an ever-widening disparity between the rate of discovery of genetic variants and our current understanding of their functions and potential roles in disease. Systematic methods for phenotyping DNA variants are required to effectively translate genomics data into improved outcomes for patients with genetic diseases. To make the biggest impact, these approaches must be scalable and accurate, faithfully reflect disease biology, and define complex disease mechanisms. We compare current methods to analyze the function of variants in their endogenous DNA context using genome editing strategies, such as saturation genome editing, base editing and prime editing. We discuss how these technologies can be linked to high-content readouts to gain deep mechanistic insights into variant effects. Finally, we highlight key challenges that need to be addressed to bridge the genotype to phenotype gap, and ultimately improve the diagnosis and treatment of genetic diseases.
基因组学的不断进步导致基因变异的发现速度与我们目前对其功能和潜在疾病作用的了解之间的差距越来越大。要想有效地将基因组学数据转化为改善遗传病患者治疗效果的方法,就必须采用系统的 DNA 变异表型分析方法。为了产生最大的影响,这些方法必须具有可扩展性和准确性,能忠实反映疾病生物学特性,并能确定复杂的疾病机制。我们比较了目前使用基因组编辑策略(如饱和基因组编辑、碱基编辑和质粒编辑)分析变体在其内源 DNA 背景下的功能的方法。我们讨论了如何将这些技术与高含量读数联系起来,以深入了解变异效应的机理。最后,我们强调了需要应对的关键挑战,以弥合基因型与表型之间的差距,最终改善遗传病的诊断和治疗。
{"title":"Analyzing the functional effects of DNA variants with gene editing.","authors":"Sarah Cooper, Sofia Obolenski, Andrew J Waters, Andrew R Bassett, Matthew A Coelho","doi":"10.1016/j.crmeth.2024.100776","DOIUrl":"10.1016/j.crmeth.2024.100776","url":null,"abstract":"<p><p>Continual advancements in genomics have led to an ever-widening disparity between the rate of discovery of genetic variants and our current understanding of their functions and potential roles in disease. Systematic methods for phenotyping DNA variants are required to effectively translate genomics data into improved outcomes for patients with genetic diseases. To make the biggest impact, these approaches must be scalable and accurate, faithfully reflect disease biology, and define complex disease mechanisms. We compare current methods to analyze the function of variants in their endogenous DNA context using genome editing strategies, such as saturation genome editing, base editing and prime editing. We discuss how these technologies can be linked to high-content readouts to gain deep mechanistic insights into variant effects. Finally, we highlight key challenges that need to be addressed to bridge the genotype to phenotype gap, and ultimately improve the diagnosis and treatment of genetic diseases.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11133854/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140923521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-20Epub Date: 2024-05-14DOI: 10.1016/j.crmeth.2024.100774
Sonali A Gandhi, Shahnaz Parveen, Munirah Alduhailan, Ramesh Tripathi, Nasser Junedi, Mohammad Saqallah, Matthew A Sanders, Peter M Hoffmann, Katherine Truex, James G Granneman, Christopher V Kelly
We present methods for making and testing the membrane biophysics of model lipid droplets (LDs). Methods are described for imaging LDs ranging in size from 0.1 to 40 μm in diameter with high-resolution microscopy and spectroscopy. With known LD compositions, membrane binding, sorting, diffusion, and tension were measured via fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging microscopy (FLIM), atomic force microscopy (AFM), and imaging flow cytometry. Additionally, a custom, small-volume pendant droplet tensiometer is described and used to measure the association of phospholipids to the LD surface. These complementary, cross-validating methods of measuring LD membrane behavior reveal the interplay of biophysical processes on lipid droplet monolayers.
{"title":"Methods for making and observing model lipid droplets.","authors":"Sonali A Gandhi, Shahnaz Parveen, Munirah Alduhailan, Ramesh Tripathi, Nasser Junedi, Mohammad Saqallah, Matthew A Sanders, Peter M Hoffmann, Katherine Truex, James G Granneman, Christopher V Kelly","doi":"10.1016/j.crmeth.2024.100774","DOIUrl":"10.1016/j.crmeth.2024.100774","url":null,"abstract":"<p><p>We present methods for making and testing the membrane biophysics of model lipid droplets (LDs). Methods are described for imaging LDs ranging in size from 0.1 to 40 μm in diameter with high-resolution microscopy and spectroscopy. With known LD compositions, membrane binding, sorting, diffusion, and tension were measured via fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging microscopy (FLIM), atomic force microscopy (AFM), and imaging flow cytometry. Additionally, a custom, small-volume pendant droplet tensiometer is described and used to measure the association of phospholipids to the LD surface. These complementary, cross-validating methods of measuring LD membrane behavior reveal the interplay of biophysical processes on lipid droplet monolayers.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11133809/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140946125","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-01DOI: 10.1016/j.crmeth.2024.100779
Peter N Nwokoye, O. Abilez
{"title":"Bioengineering methods for vascularizing organoids.","authors":"Peter N Nwokoye, O. Abilez","doi":"10.1016/j.crmeth.2024.100779","DOIUrl":"https://doi.org/10.1016/j.crmeth.2024.100779","url":null,"abstract":"","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141030678","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-04-22Epub Date: 2024-03-29DOI: 10.1016/j.crmeth.2024.100739
Anja R Köhler, Johannes Haußer, Annika Harsch, Steffen Bernhardt, Lilia Häußermann, Lisa-Marie Brenner, Cristiana Lungu, Monilola A Olayioye, Pavel Bashtrykov, Albert Jeltsch
Dynamic changes in the epigenome at defined genomic loci play crucial roles during cellular differentiation and disease development. Here, we developed dual-color bimolecular anchor detector (BiAD) sensors for high-sensitivity readout of locus-specific epigenome modifications by fluorescence microscopy. Our BiAD sensors comprise an sgRNA/dCas9 complex as anchor and double chromatin reader domains as detector modules, both fused to complementary parts of a split IFP2.0 fluorophore, enabling its reconstitution upon binding of both parts in close proximity. In addition, a YPet fluorophore is recruited to the sgRNA to mark the genomic locus of interest. With these dual-color BiAD sensors, we detected H3K9me2/3 and DNA methylation and their dynamic changes upon RNAi or inhibitor treatment with high sensitivity at endogenous genomic regions. Furthermore, we showcased locus-specific H3K36me2/3 readout as well as H3K27me3 and H3K9me2/3 enrichment on the inactive X chromosome, highlighting the broad applicability of our dual-color BiAD sensors for single-cell epigenome studies.
{"title":"Modular dual-color BiAD sensors for locus-specific readout of epigenome modifications in single cells.","authors":"Anja R Köhler, Johannes Haußer, Annika Harsch, Steffen Bernhardt, Lilia Häußermann, Lisa-Marie Brenner, Cristiana Lungu, Monilola A Olayioye, Pavel Bashtrykov, Albert Jeltsch","doi":"10.1016/j.crmeth.2024.100739","DOIUrl":"10.1016/j.crmeth.2024.100739","url":null,"abstract":"<p><p>Dynamic changes in the epigenome at defined genomic loci play crucial roles during cellular differentiation and disease development. Here, we developed dual-color bimolecular anchor detector (BiAD) sensors for high-sensitivity readout of locus-specific epigenome modifications by fluorescence microscopy. Our BiAD sensors comprise an sgRNA/dCas9 complex as anchor and double chromatin reader domains as detector modules, both fused to complementary parts of a split IFP2.0 fluorophore, enabling its reconstitution upon binding of both parts in close proximity. In addition, a YPet fluorophore is recruited to the sgRNA to mark the genomic locus of interest. With these dual-color BiAD sensors, we detected H3K9me2/3 and DNA methylation and their dynamic changes upon RNAi or inhibitor treatment with high sensitivity at endogenous genomic regions. Furthermore, we showcased locus-specific H3K36me2/3 readout as well as H3K27me3 and H3K9me2/3 enrichment on the inactive X chromosome, highlighting the broad applicability of our dual-color BiAD sensors for single-cell epigenome studies.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11045877/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140330147","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The pathogenesis of Alzheimer disease (AD) involves complex gene regulatory changes across different cell types. To help decipher this complexity, we introduce single-cell Bayesian biclustering (scBC), a framework for identifying cell-specific gene network biomarkers in scRNA and snRNA-seq data. Through biclustering, scBC enables the analysis of perturbations in functional gene modules at the single-cell level. Applying the scBC framework to AD snRNA-seq data reveals the perturbations within gene modules across distinct cell groups and sheds light on gene-cell correlations during AD progression. Notably, our method helps to overcome common challenges in single-cell data analysis, including batch effects and dropout events. Incorporating prior knowledge further enables the framework to yield more biologically interpretable results. Comparative analyses on simulated and real-world datasets demonstrate the precision and robustness of our approach compared to other state-of-the-art biclustering methods. scBC holds potential for unraveling the mechanisms underlying polygenic diseases characterized by intricate gene coexpression patterns.
阿尔茨海默病(AD)的发病机制涉及不同细胞类型的复杂基因调控变化。为了帮助破译这种复杂性,我们引入了单细胞贝叶斯双聚类(scBC),这是一种在 scRNA 和 snRNA-seq 数据中识别细胞特异性基因网络生物标记物的框架。通过双聚类,scBC 能够在单细胞水平上分析功能基因模块的扰动。将 scBC 框架应用于 AD snRNA-seq 数据可揭示不同细胞群中基因模块的扰动,并揭示 AD 进展过程中基因与细胞的相关性。值得注意的是,我们的方法有助于克服单细胞数据分析中常见的挑战,包括批次效应和丢失事件。结合先验知识还能使该框架产生更具生物学解释性的结果。对模拟数据集和真实世界数据集的比较分析表明,与其他最先进的双聚类方法相比,我们的方法既精确又稳健。
{"title":"Single-cell biclustering for cell-specific transcriptomic perturbation detection in AD progression.","authors":"Yuqiao Gong, Jingsi Xu, Maoying Wu, Ruitian Gao, Jianle Sun, Zhangsheng Yu, Yue Zhang","doi":"10.1016/j.crmeth.2024.100742","DOIUrl":"10.1016/j.crmeth.2024.100742","url":null,"abstract":"<p><p>The pathogenesis of Alzheimer disease (AD) involves complex gene regulatory changes across different cell types. To help decipher this complexity, we introduce single-cell Bayesian biclustering (scBC), a framework for identifying cell-specific gene network biomarkers in scRNA and snRNA-seq data. Through biclustering, scBC enables the analysis of perturbations in functional gene modules at the single-cell level. Applying the scBC framework to AD snRNA-seq data reveals the perturbations within gene modules across distinct cell groups and sheds light on gene-cell correlations during AD progression. Notably, our method helps to overcome common challenges in single-cell data analysis, including batch effects and dropout events. Incorporating prior knowledge further enables the framework to yield more biologically interpretable results. Comparative analyses on simulated and real-world datasets demonstrate the precision and robustness of our approach compared to other state-of-the-art biclustering methods. scBC holds potential for unraveling the mechanisms underlying polygenic diseases characterized by intricate gene coexpression patterns.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11045878/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140330149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-04-22Epub Date: 2024-03-15DOI: 10.1016/j.crmeth.2024.100728
Markus Dobersberger, Delia Sumesgutner, Charlotte U Zajc, Benjamin Salzer, Elisabeth Laurent, Dominik Emminger, Elise Sylvander, Elisabeth Lehner, Magdalena Teufl, Jacqueline Seigner, Madhusudhan Reddy Bobbili, Renate Kunert, Manfred Lehner, Michael W Traxlmayr
Chimeric antigen receptor (CAR) T cells have shown remarkable response rates in hematological malignancies. In contrast, CAR T cell treatment of solid tumors is associated with several challenges, in particular the expression of most tumor-associated antigens at lower levels in vital organs, resulting in on-target/off-tumor toxicities. Thus, innovative approaches to improve the tumor specificity of CAR T cells are urgently needed. Based on the observation that many human solid tumors activate epidermal growth factor receptor (EGFR) on their surface through secretion of EGFR ligands, we developed an engineering strategy for CAR-binding domains specifically directed against the ligand-activated conformation of EGFR. We show, in several experimental systems, that the generated binding domains indeed enable CAR T cells to distinguish between active and inactive EGFR. We anticipate that this engineering concept will be an important step forward to improve the tumor specificity of CAR T cells directed against EGFR-positive solid cancers.
嵌合抗原受体(CAR)T 细胞在血液恶性肿瘤中显示出显著的反应率。相比之下,CAR T 细胞治疗实体瘤却面临着一些挑战,特别是大多数肿瘤相关抗原在重要器官中的表达水平较低,导致靶上/非肿瘤毒性。因此,迫切需要创新方法来提高 CAR T 细胞的肿瘤特异性。根据对许多人类实体瘤通过分泌表皮生长因子受体配体激活其表面的表皮生长因子受体(EGFR)的观察,我们开发了一种工程策略,专门针对表皮生长因子受体配体激活构象的 CAR 结合域。我们在多个实验系统中证明,生成的结合域确实能使 CAR T 细胞区分表皮生长因子受体的活性和非活性。我们预计,这一工程概念将是提高 CAR T 细胞针对表皮生长因子受体阳性实体癌的肿瘤特异性的重要一步。
{"title":"An engineering strategy to target activated EGFR with CAR T cells.","authors":"Markus Dobersberger, Delia Sumesgutner, Charlotte U Zajc, Benjamin Salzer, Elisabeth Laurent, Dominik Emminger, Elise Sylvander, Elisabeth Lehner, Magdalena Teufl, Jacqueline Seigner, Madhusudhan Reddy Bobbili, Renate Kunert, Manfred Lehner, Michael W Traxlmayr","doi":"10.1016/j.crmeth.2024.100728","DOIUrl":"10.1016/j.crmeth.2024.100728","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T cells have shown remarkable response rates in hematological malignancies. In contrast, CAR T cell treatment of solid tumors is associated with several challenges, in particular the expression of most tumor-associated antigens at lower levels in vital organs, resulting in on-target/off-tumor toxicities. Thus, innovative approaches to improve the tumor specificity of CAR T cells are urgently needed. Based on the observation that many human solid tumors activate epidermal growth factor receptor (EGFR) on their surface through secretion of EGFR ligands, we developed an engineering strategy for CAR-binding domains specifically directed against the ligand-activated conformation of EGFR. We show, in several experimental systems, that the generated binding domains indeed enable CAR T cells to distinguish between active and inactive EGFR. We anticipate that this engineering concept will be an important step forward to improve the tumor specificity of CAR T cells directed against EGFR-positive solid cancers.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":3.8,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11045874/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140140902","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-04-22Epub Date: 2024-03-29DOI: 10.1016/j.crmeth.2024.100743
Camila P Camargo, Yunus Alapan, Abir K Muhuri, Samuel N Lucas, Susan N Thomas
Tissue infiltration by circulating leukocytes occurs via adhesive interactions with the local vasculature, but how the adhesive quality of circulating cells guides the homing of specific phenotypes to different vascular microenvironments remains undefined. We developed an optofluidic system enabling fluorescent labeling of photoactivatable cells based on their adhesive rolling velocity in an inflamed vasculature-mimicking microfluidic device under physiological fluid flow. In so doing, single-cell level multidimensional profiling of cellular characteristics could be characterized and related to the associated adhesive phenotype. When applied to CD8+ T cells, ligand/receptor expression profiles and subtypes associated with adhesion were revealed, providing insight into inflamed tissue infiltration capabilities of specific CD8+ T lymphocyte subsets and how local vascular microenvironmental features may regulate the quality of cellular infiltration. This methodology facilitates rapid screening of cell populations for enhanced homing capabilities under defined biochemical and biophysical microenvironments, relevant to leukocyte homing modulation in multiple pathologies.
循环白细胞通过与局部血管的粘附相互作用进行组织浸润,但循环细胞的粘附质量如何引导特定表型的细胞向不同的血管微环境归巢仍未确定。我们开发了一种光流体系统,可根据细胞在生理流体流动下的炎症血管模拟微流体装置中的粘附滚动速度,对光激活细胞进行荧光标记。这样就能在单细胞水平上对细胞特征进行多维剖析,并将其与相关的粘附表型联系起来。当应用于 CD8+ T 细胞时,与粘附相关的配体/受体表达谱和亚型被揭示出来,让人们深入了解特定 CD8+ T 淋巴细胞亚群的炎症组织浸润能力,以及局部血管微环境特征如何调节细胞浸润的质量。这种方法有助于在确定的生化和生物物理微环境下快速筛选增强归巢能力的细胞群,这与多种病症中的白细胞归巢调节有关。
{"title":"Single-cell adhesive profiling in an optofluidic device elucidates CD8<sup>+</sup> T lymphocyte phenotypes in inflamed vasculature-like microenvironments.","authors":"Camila P Camargo, Yunus Alapan, Abir K Muhuri, Samuel N Lucas, Susan N Thomas","doi":"10.1016/j.crmeth.2024.100743","DOIUrl":"10.1016/j.crmeth.2024.100743","url":null,"abstract":"<p><p>Tissue infiltration by circulating leukocytes occurs via adhesive interactions with the local vasculature, but how the adhesive quality of circulating cells guides the homing of specific phenotypes to different vascular microenvironments remains undefined. We developed an optofluidic system enabling fluorescent labeling of photoactivatable cells based on their adhesive rolling velocity in an inflamed vasculature-mimicking microfluidic device under physiological fluid flow. In so doing, single-cell level multidimensional profiling of cellular characteristics could be characterized and related to the associated adhesive phenotype. When applied to CD8<sup>+</sup> T cells, ligand/receptor expression profiles and subtypes associated with adhesion were revealed, providing insight into inflamed tissue infiltration capabilities of specific CD8<sup>+</sup> T lymphocyte subsets and how local vascular microenvironmental features may regulate the quality of cellular infiltration. This methodology facilitates rapid screening of cell populations for enhanced homing capabilities under defined biochemical and biophysical microenvironments, relevant to leukocyte homing modulation in multiple pathologies.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11046032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140330148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}