Pub Date : 2025-10-20Epub Date: 2025-09-18DOI: 10.1016/j.crmeth.2025.101182
Moritz Becker, Nahid Safari, Christian Tetzlaff
Recent advances in molecular biology have led to large-scale datasets providing new insights into the molecular organization of cells. To fully exploit their potential, computer simulations are essential to gain in-depth understanding of molecular principles. We developed the Python reaction interaction diffusion simulator (PyRID), a Python-based reaction-diffusion simulator designed for the efficient simulation of molecular biological systems. PyRID incorporates unimolecular and bimolecular reactions as well as pair interactions and simulation of individual interacting proteins to polydisperse molecular assemblies. It supports mesh-based compartments and surface diffusion of particles, enabling analyses of interactions between (trans)membrane proteins with intra- and extracellular proteins. Distinctively, PyRID uses hierarchical grids for polydisperse systems, supports rigid bead models, and calculates diffusion tensors internally. Validation against theoretical results and established models confirms PyRID's accuracy in reproducing key physical properties. PyRID is written entirely in Python, making it accessible to the broader scientific community, facilitating customization and integration into diverse research workflows.
{"title":"The Brownian dynamics simulator PyRID for reacting and interacting particles written in Python.","authors":"Moritz Becker, Nahid Safari, Christian Tetzlaff","doi":"10.1016/j.crmeth.2025.101182","DOIUrl":"10.1016/j.crmeth.2025.101182","url":null,"abstract":"<p><p>Recent advances in molecular biology have led to large-scale datasets providing new insights into the molecular organization of cells. To fully exploit their potential, computer simulations are essential to gain in-depth understanding of molecular principles. We developed the Python reaction interaction diffusion simulator (PyRID), a Python-based reaction-diffusion simulator designed for the efficient simulation of molecular biological systems. PyRID incorporates unimolecular and bimolecular reactions as well as pair interactions and simulation of individual interacting proteins to polydisperse molecular assemblies. It supports mesh-based compartments and surface diffusion of particles, enabling analyses of interactions between (trans)membrane proteins with intra- and extracellular proteins. Distinctively, PyRID uses hierarchical grids for polydisperse systems, supports rigid bead models, and calculates diffusion tensors internally. Validation against theoretical results and established models confirms PyRID's accuracy in reproducing key physical properties. PyRID is written entirely in Python, making it accessible to the broader scientific community, facilitating customization and integration into diverse research workflows.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101182"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570352/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092547","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 : 2025-10-20Epub Date: 2025-09-19DOI: 10.1016/j.crmeth.2025.101183
Xu-Wen Wang, Min Hyung Ryu, Michael H Cho, Peter Castaldi, Craig P Hersh, Edwin K Silverman, Yang-Yu Liu
Genes associated with the same disease frequently engage in mutual biological interactions, e.g., perturbation within a specific neighborhood in the molecular interactome, often referred to as the disease module. This has propelled the advancement of network-based approaches toward elucidating the molecular bases of human diseases. Although many computational methods have been developed to integrate the molecular interactome and omics profiles to extract such context-dependent disease modules, approaches that leverage multi-omics for disease-module detection are still lacking. Here, we developed a statistical physics approach based on the random-field O(n) model (RFOnM) to fill this gap. We applied the RFOnM approach to integrate gene-expression data and genome-wide association studies or mRNA data and DNA methylation for several complex diseases with the human interactome. We found that the RFOnM approach outperforms existing single omics methods in most of the complex diseases considered in this study.
{"title":"A statistical physics approach to integrating multi-omics data for disease-module detection.","authors":"Xu-Wen Wang, Min Hyung Ryu, Michael H Cho, Peter Castaldi, Craig P Hersh, Edwin K Silverman, Yang-Yu Liu","doi":"10.1016/j.crmeth.2025.101183","DOIUrl":"10.1016/j.crmeth.2025.101183","url":null,"abstract":"<p><p>Genes associated with the same disease frequently engage in mutual biological interactions, e.g., perturbation within a specific neighborhood in the molecular interactome, often referred to as the disease module. This has propelled the advancement of network-based approaches toward elucidating the molecular bases of human diseases. Although many computational methods have been developed to integrate the molecular interactome and omics profiles to extract such context-dependent disease modules, approaches that leverage multi-omics for disease-module detection are still lacking. Here, we developed a statistical physics approach based on the random-field O(n) model (RFOnM) to fill this gap. We applied the RFOnM approach to integrate gene-expression data and genome-wide association studies or mRNA data and DNA methylation for several complex diseases with the human interactome. We found that the RFOnM approach outperforms existing single omics methods in most of the complex diseases considered in this study.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101183"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570349/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145103040","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 : 2025-10-20Epub Date: 2025-10-14DOI: 10.1016/j.crmeth.2025.101203
Santiago Solé-Domènech, Pradeep Kumar Singh, Lucy Funes, Cheng-I J Ma, J David Warren, Frederick R Maxfield
Active endolysosomal pH regulation is essential for optimal enzymatic activity. To measure acidification, pH sensors can be delivered to acidic compartments using labeled dextran polymers or proteins. However, commercial probes have limited sensitivity in the acidic endolysosomal range or their fluorescence undergoes degradation. Herein, we introduce the new pH-sensitive probe ApHID, a green-emitting sensor with optimal dynamic range matching the acidity of endosomes and lysosomes. Acid pH indicator dye (ApHID) has a pKa near 5, increasing brightness with acidity, and withstands oxidation and photobleaching. We used ApHID dextrans to measure endolysosomal pH in macrophages and compared it to other commercially available sensors. ApHID reported pH accurately and stably over time in cell culture and was sensitive to subtle variations in organelle acidification in real time. Overall, ApHID circumvents limitations of currently available commercial probes and can provide utility in demanding applications such as intravital imaging of tissues.
{"title":"Real-time pH imaging of macrophage lysosomes using the pH-sensitive probe ApHID.","authors":"Santiago Solé-Domènech, Pradeep Kumar Singh, Lucy Funes, Cheng-I J Ma, J David Warren, Frederick R Maxfield","doi":"10.1016/j.crmeth.2025.101203","DOIUrl":"10.1016/j.crmeth.2025.101203","url":null,"abstract":"<p><p>Active endolysosomal pH regulation is essential for optimal enzymatic activity. To measure acidification, pH sensors can be delivered to acidic compartments using labeled dextran polymers or proteins. However, commercial probes have limited sensitivity in the acidic endolysosomal range or their fluorescence undergoes degradation. Herein, we introduce the new pH-sensitive probe ApHID, a green-emitting sensor with optimal dynamic range matching the acidity of endosomes and lysosomes. Acid pH indicator dye (ApHID) has a pKa near 5, increasing brightness with acidity, and withstands oxidation and photobleaching. We used ApHID dextrans to measure endolysosomal pH in macrophages and compared it to other commercially available sensors. ApHID reported pH accurately and stably over time in cell culture and was sensitive to subtle variations in organelle acidification in real time. Overall, ApHID circumvents limitations of currently available commercial probes and can provide utility in demanding applications such as intravital imaging of tissues.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101203"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570321/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145303673","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 : 2025-10-20Epub Date: 2025-09-15DOI: 10.1016/j.crmeth.2025.101175
Farhan Ameen, Nick Robertson, David M Lin, Shila Ghazanfar, Ellis Patrick
Spatial proteomic and transcriptomic technologies enable high-throughput phenotyping of cells in situ, enabling quantification of spatial relationships among diverse cell populations. However, the experimental design choice of which regions of a tissue will be imaged can greatly impact the interpretation of spatial quantifications. That is, spatial relationships identified in one region of interest may not be interpreted consistently across other regions. To address this challenge, we introduce Kontextual, a method that considers alternative frames of reference for contextualizing spatial relationships. These contexts may represent landmarks, spatial domains, or groups of functionally similar cells that are consistent across regions. By modeling spatial relationships between cells relative to these contexts, Kontextual produces robust spatial quantifications that are not confounded by the region selected. We demonstrate in spatial proteomics and transcriptomics datasets that modeling spatial relationships this way is biologically meaningful and can improve the prediction of patient prognosis in a classification setting.
{"title":"Kontextual reframes analysis of spatial omics data and reveals consistent cell relationships across images.","authors":"Farhan Ameen, Nick Robertson, David M Lin, Shila Ghazanfar, Ellis Patrick","doi":"10.1016/j.crmeth.2025.101175","DOIUrl":"10.1016/j.crmeth.2025.101175","url":null,"abstract":"<p><p>Spatial proteomic and transcriptomic technologies enable high-throughput phenotyping of cells in situ, enabling quantification of spatial relationships among diverse cell populations. However, the experimental design choice of which regions of a tissue will be imaged can greatly impact the interpretation of spatial quantifications. That is, spatial relationships identified in one region of interest may not be interpreted consistently across other regions. To address this challenge, we introduce Kontextual, a method that considers alternative frames of reference for contextualizing spatial relationships. These contexts may represent landmarks, spatial domains, or groups of functionally similar cells that are consistent across regions. By modeling spatial relationships between cells relative to these contexts, Kontextual produces robust spatial quantifications that are not confounded by the region selected. We demonstrate in spatial proteomics and transcriptomics datasets that modeling spatial relationships this way is biologically meaningful and can improve the prediction of patient prognosis in a classification setting.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101175"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570319/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145076260","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 : 2025-10-20Epub Date: 2025-09-24DOI: 10.1016/j.crmeth.2025.101184
Xiaoqing Ru, Lifeng Xu, Wu Han, Quan Zou
Drug-target interaction (DTI) prediction is a crucial component of drug discovery. In recent years, in silico approaches have attracted attention for DTI prediction, primarily because of their potential to mitigate the high costs, low success rates, and extensive timelines of traditional drug development, while efficiently using the growing amount of available data. This review identifies four major factors that influence DTI predictions, highlights persistent challenges, and proposes insights and strategies from the perspectives of data, features, and experimental setups to address these challenges. Furthermore, it emphasizes the importance of refining established approaches-such as the "guilt-by-association" concept-to manage data sparsity, and integrating emerging technologies, including large language models and AlphaFold, to advance feature engineering. We hope that this work will provide valuable guidance and novel perspectives for advancing future research on DTI predictions.
{"title":"In silico methods for drug-target interaction prediction.","authors":"Xiaoqing Ru, Lifeng Xu, Wu Han, Quan Zou","doi":"10.1016/j.crmeth.2025.101184","DOIUrl":"10.1016/j.crmeth.2025.101184","url":null,"abstract":"<p><p>Drug-target interaction (DTI) prediction is a crucial component of drug discovery. In recent years, in silico approaches have attracted attention for DTI prediction, primarily because of their potential to mitigate the high costs, low success rates, and extensive timelines of traditional drug development, while efficiently using the growing amount of available data. This review identifies four major factors that influence DTI predictions, highlights persistent challenges, and proposes insights and strategies from the perspectives of data, features, and experimental setups to address these challenges. Furthermore, it emphasizes the importance of refining established approaches-such as the \"guilt-by-association\" concept-to manage data sparsity, and integrating emerging technologies, including large language models and AlphaFold, to advance feature engineering. We hope that this work will provide valuable guidance and novel perspectives for advancing future research on DTI predictions.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101184"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570315/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145151024","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 : 2025-10-20Epub Date: 2025-09-17DOI: 10.1016/j.crmeth.2025.101178
Yusuke Imoto
Single-cell sequencing enables genome- and epigenome-wide profiling of thousands of individual cells, offering unprecedented biological insights. However, technical noise and batch effects obscure high-resolution structures, hindering rare-cell-type detection and cross-dataset comparisons. To comprehensively address these challenges, this study upgrades RECODE, a high-dimensional statistics-based tool for technical noise reduction in single-cell RNA sequencing (RNA-seq), to include a function called iRECODE, which simultaneously reduces technical and batch noise. Further, RECODE's applicability is extended to diverse single-cell modalities, including single-cell high-throughput chromosome conformation capture (Hi-C) and spatial transcriptomics. Recent improvements in the algorithm have substantially enhanced both accuracy and computational efficiency. The RECODE platform thus provides a robust and versatile solution for noise mitigation, enabling more accurate downstream analyses across transcriptomic, epigenomic, and spatial domains.
{"title":"Comprehensive noise reduction in single-cell data with the RECODE platform.","authors":"Yusuke Imoto","doi":"10.1016/j.crmeth.2025.101178","DOIUrl":"10.1016/j.crmeth.2025.101178","url":null,"abstract":"<p><p>Single-cell sequencing enables genome- and epigenome-wide profiling of thousands of individual cells, offering unprecedented biological insights. However, technical noise and batch effects obscure high-resolution structures, hindering rare-cell-type detection and cross-dataset comparisons. To comprehensively address these challenges, this study upgrades RECODE, a high-dimensional statistics-based tool for technical noise reduction in single-cell RNA sequencing (RNA-seq), to include a function called iRECODE, which simultaneously reduces technical and batch noise. Further, RECODE's applicability is extended to diverse single-cell modalities, including single-cell high-throughput chromosome conformation capture (Hi-C) and spatial transcriptomics. Recent improvements in the algorithm have substantially enhanced both accuracy and computational efficiency. The RECODE platform thus provides a robust and versatile solution for noise mitigation, enabling more accurate downstream analyses across transcriptomic, epigenomic, and spatial domains.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101178"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570323/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145087446","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 : 2025-10-20Epub Date: 2025-10-14DOI: 10.1016/j.crmeth.2025.101202
Lucy Liang, Alessandro Fasse, Arianna Damiani, Maria K Jantz, Uzoma Agbor, Matteo Del Brocco, Rachel Monney, Cecelia Rowland, Taylor Newton, Chaitanya Gopinath, David J Schaeffer, Christi L Kolarcik, John G Pagiazitis, George Z Mentis, Lee E Fisher, Esra Neufeld, Marco Capogrosso, Robert A Gaunt, T Kevin Hitchens, Elvira Pirondini
High-definition visualization techniques are critical for understanding the neuroanatomy of the spinal cord, an essential structure for sensorimotor and autonomic functions, in both healthy and pathological conditions. Magnetic resonance imaging (MRI) is a common method for visualizing neural structures in 3D. However, techniques for spinal cord MRI have historically achieved limited visualization of rootlets and nerves, especially at lower spinal levels, due to their highly complex and compact organization. Here, we developed a spinal in situ contrast 3D imaging (SpIC3D) method that allows visualization of spinal compartments in fixed animal and human specimens with high resolution (50 μm) at various spinal levels. Using SpIC3D, we achieved quantification of neuronal cell density in dorsal root ganglia, multi-segment identification of individual rootlets and roots, and volumetric reconstruction of multiple spinal structures for computational modeling. SpIC3D provides a basis for accelerated spinal pathology characterization and personalized spinal cord stimulation treatments.
{"title":"SpIC3D imaging for spinal in situ contrast 3D visualization.","authors":"Lucy Liang, Alessandro Fasse, Arianna Damiani, Maria K Jantz, Uzoma Agbor, Matteo Del Brocco, Rachel Monney, Cecelia Rowland, Taylor Newton, Chaitanya Gopinath, David J Schaeffer, Christi L Kolarcik, John G Pagiazitis, George Z Mentis, Lee E Fisher, Esra Neufeld, Marco Capogrosso, Robert A Gaunt, T Kevin Hitchens, Elvira Pirondini","doi":"10.1016/j.crmeth.2025.101202","DOIUrl":"10.1016/j.crmeth.2025.101202","url":null,"abstract":"<p><p>High-definition visualization techniques are critical for understanding the neuroanatomy of the spinal cord, an essential structure for sensorimotor and autonomic functions, in both healthy and pathological conditions. Magnetic resonance imaging (MRI) is a common method for visualizing neural structures in 3D. However, techniques for spinal cord MRI have historically achieved limited visualization of rootlets and nerves, especially at lower spinal levels, due to their highly complex and compact organization. Here, we developed a spinal in situ contrast 3D imaging (SpIC3D) method that allows visualization of spinal compartments in fixed animal and human specimens with high resolution (50 μm) at various spinal levels. Using SpIC3D, we achieved quantification of neuronal cell density in dorsal root ganglia, multi-segment identification of individual rootlets and roots, and volumetric reconstruction of multiple spinal structures for computational modeling. SpIC3D provides a basis for accelerated spinal pathology characterization and personalized spinal cord stimulation treatments.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101202"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145303724","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 : 2025-10-20Epub Date: 2025-09-17DOI: 10.1016/j.crmeth.2025.101176
Yan Wang, Charlotte van Dijk, Ilia Timpanaro, Paul Hop, Brendan Kenna, Maarten Kooyman, Eleonora Aronica, R Jeroen Pasterkamp, Leonard H van den Berg, Johnathan Cooper-Knock, Jan H Veldink, Kevin Kenna
We developed SpliPath as a generalizable framework to discover disease associations mediated by rare variants that induce experimentally supported mRNA splicing defects. Our approach integrates components of burden tests (BTs), traditional splicing quantitative trait locus (sQTL) analyses, and sequence-to-function AI models (SpliceAI and Pangolin). Central to the workings of SpliPath is our concept of collapsed rare variant splicing QTL (crsQTL). crsQTL groups rare variants that are predicted to alter splicing in the same way, specifically by linking them to shared splice junctions observed in independent (unpaired) RNA sequencing (RNA-seq) datasets. We demonstrate the utility of SpliPath through applications in amyotrophic lateral sclerosis (ALS). Through this, we showcase scenarios where SpliPath detects genetic associations that cannot be recovered by more simplistic combinations of BT and SpliceAI. We also nominate crsQTL for splice defects detected in large-scale analyses of ALS patient tissue.
{"title":"SpliPath enhances disease gene discovery in case-control analyses of rare splice-altering genetic variants.","authors":"Yan Wang, Charlotte van Dijk, Ilia Timpanaro, Paul Hop, Brendan Kenna, Maarten Kooyman, Eleonora Aronica, R Jeroen Pasterkamp, Leonard H van den Berg, Johnathan Cooper-Knock, Jan H Veldink, Kevin Kenna","doi":"10.1016/j.crmeth.2025.101176","DOIUrl":"10.1016/j.crmeth.2025.101176","url":null,"abstract":"<p><p>We developed SpliPath as a generalizable framework to discover disease associations mediated by rare variants that induce experimentally supported mRNA splicing defects. Our approach integrates components of burden tests (BTs), traditional splicing quantitative trait locus (sQTL) analyses, and sequence-to-function AI models (SpliceAI and Pangolin). Central to the workings of SpliPath is our concept of collapsed rare variant splicing QTL (crsQTL). crsQTL groups rare variants that are predicted to alter splicing in the same way, specifically by linking them to shared splice junctions observed in independent (unpaired) RNA sequencing (RNA-seq) datasets. We demonstrate the utility of SpliPath through applications in amyotrophic lateral sclerosis (ALS). Through this, we showcase scenarios where SpliPath detects genetic associations that cannot be recovered by more simplistic combinations of BT and SpliceAI. We also nominate crsQTL for splice defects detected in large-scale analyses of ALS patient tissue.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101176"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570325/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145087405","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 : 2025-10-20Epub Date: 2025-09-17DOI: 10.1016/j.crmeth.2025.101177
Negin Imani Farahani, Kenneth Kin Lam Wong, George Allen, Abhimanyu Minhas, Lisa Lin, Shama Nazir, Lisa M Julian
In vitro human pluripotent stem cell (hPSC)-derived models have been crucial in advancing our understanding of the mechanisms underlying neurodevelopment, though knowledge of the earliest stages of brain formation is lacking. Micropatterning of cell populations as they transition from pluripotency through the process of neurulation can produce self-assembled neuroepithelial tissues (NETs) with precise spatiotemporal control, enhancing the fidelity of hPSC models to the early developing human brain and their use in phenotypic assessments. Here, we introduce an accessible, customizable, and scalable method to produce self-assembled NETs using bioprinting to rapidly deposit reproducibly sized extracellular matrix droplets. Matrix addition to the media provides a scaffold that promotes 3D tissue folding, reflecting neural tube development. We demonstrate that these scaffolded NETs (scNETs) exhibit key architectural and biological features of the human brain during normal and abnormal development-notably, hyperproliferation and structural malformations induced by TSC2 deficiency-and provide a robust drug screening tool.
{"title":"High-throughput bioprinting to produce micropatterned neuroepithelial tissues and model TSC2-deficient brain malformations.","authors":"Negin Imani Farahani, Kenneth Kin Lam Wong, George Allen, Abhimanyu Minhas, Lisa Lin, Shama Nazir, Lisa M Julian","doi":"10.1016/j.crmeth.2025.101177","DOIUrl":"10.1016/j.crmeth.2025.101177","url":null,"abstract":"<p><p>In vitro human pluripotent stem cell (hPSC)-derived models have been crucial in advancing our understanding of the mechanisms underlying neurodevelopment, though knowledge of the earliest stages of brain formation is lacking. Micropatterning of cell populations as they transition from pluripotency through the process of neurulation can produce self-assembled neuroepithelial tissues (NETs) with precise spatiotemporal control, enhancing the fidelity of hPSC models to the early developing human brain and their use in phenotypic assessments. Here, we introduce an accessible, customizable, and scalable method to produce self-assembled NETs using bioprinting to rapidly deposit reproducibly sized extracellular matrix droplets. Matrix addition to the media provides a scaffold that promotes 3D tissue folding, reflecting neural tube development. We demonstrate that these scaffolded NETs (scNETs) exhibit key architectural and biological features of the human brain during normal and abnormal development-notably, hyperproliferation and structural malformations induced by TSC2 deficiency-and provide a robust drug screening tool.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101177"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570318/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145087422","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 : 2025-10-20Epub Date: 2025-09-26DOI: 10.1016/j.crmeth.2025.101185
Nils Mülling, J Fréderique de Graaf, Graham A Heieis, Kristina Boss, Benjamin Wilde, Bart Everts, Ramon Arens
Cytotoxic CD8+ T cells are essential mediators of immune responses against viral infections and tumors. Upon antigen encounter, antigen-specific CD8+ T cells undergo clonal expansion and produce effector cytokines, processes that require dynamic metabolic adaptation. However, profiling antigen-specific T cells at single-cell resolution remains technically challenging. We present a spectral flow cytometry-based workflow enabling metabolic profiling of antigen-specific CD8+ T cells identified via major histocompatibility complex (MHC) class I tetramers or CD137 upregulation. The approach integrates the analysis of metabolic protein expression to infer pathway activity, uptake of fluorescent probes to measure functional metabolism and metabolite utilization, and assays evaluating cellular energy metabolism. Applied to human and mouse samples, this method defined the metabolic profiles of cytomegalovirus-, SARS-CoV-2-, and tumor-specific CD8+ T cells across distinct activation states and tissues. By detailing each component of the workflow, we provide practical guidance for applying metabolic spectral flow cytometry to dissect disease mechanisms and therapeutic responses.
{"title":"Metabolic profiling of antigen-specific CD8<sup>+</sup> T cells by spectral flow cytometry.","authors":"Nils Mülling, J Fréderique de Graaf, Graham A Heieis, Kristina Boss, Benjamin Wilde, Bart Everts, Ramon Arens","doi":"10.1016/j.crmeth.2025.101185","DOIUrl":"10.1016/j.crmeth.2025.101185","url":null,"abstract":"<p><p>Cytotoxic CD8<sup>+</sup> T cells are essential mediators of immune responses against viral infections and tumors. Upon antigen encounter, antigen-specific CD8<sup>+</sup> T cells undergo clonal expansion and produce effector cytokines, processes that require dynamic metabolic adaptation. However, profiling antigen-specific T cells at single-cell resolution remains technically challenging. We present a spectral flow cytometry-based workflow enabling metabolic profiling of antigen-specific CD8<sup>+</sup> T cells identified via major histocompatibility complex (MHC) class I tetramers or CD137 upregulation. The approach integrates the analysis of metabolic protein expression to infer pathway activity, uptake of fluorescent probes to measure functional metabolism and metabolite utilization, and assays evaluating cellular energy metabolism. Applied to human and mouse samples, this method defined the metabolic profiles of cytomegalovirus-, SARS-CoV-2-, and tumor-specific CD8<sup>+</sup> T cells across distinct activation states and tissues. By detailing each component of the workflow, we provide practical guidance for applying metabolic spectral flow cytometry to dissect disease mechanisms and therapeutic responses.</p>","PeriodicalId":29773,"journal":{"name":"Cell Reports Methods","volume":" ","pages":"101185"},"PeriodicalIF":4.5,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12570331/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145182218","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}