+
Improve Research Reproducibility A Bio-protocol resource

Cell Biology


Categories

Protocols in Current Issue
0 Q&A 272 Views Nov 5, 2025

When plants undergo senescence or experience carbon starvation, leaf cells degrade proteins in the chloroplasts on a massive scale via autophagy, an evolutionarily conserved process in which intracellular components are transported to the vacuole for degradation to facilitate nutrient recycling. Nonetheless, how portions of chloroplasts are released from the main chloroplast body and mobilized to the vacuole remains unclear. Here, we developed a method to observe the autophagic transport of chloroplast proteins in real time using confocal laser-scanning microscopy on transgenic plants expressing fluorescently labeled chloroplast components and autophagy-associated membranes. This protocol enabled us to track changes in chloroplast morphology during chloroplast-targeted autophagy on a timescale of seconds, and it could be adapted to monitor the dynamics of other intracellular processes in plant leaves.

0 Q&A 171 Views Nov 5, 2025

Nowadays, recombinant proteins are the focus of various research fields, and their use ranges from therapeutic investigations to cellular model systems for the development of therapeutic approaches. Cell systems used for the expression of recombinant proteins should be comparable in terms of yield and expression efficiency. In many research fields, it is desirable to obtain high protein concentrations. A method that combines an easy workflow with rapid results and affordable costs remains missing, and a standardized approach to determining protein concentration in transgenic cell lines is essential for more reliable data analysis. Our protocol demonstrates the cluster fluorescence-linked immunosorbent assay (FLISA), a technique that allows the exact quantification of comparable protein expression amounts. Moreover, it enables the detection of clustered or bound subunits of a protein without necessitating ultracentrifugation. In the present protocol, we demonstrate the utilization of two transgene cell lines, each expressing distinct recombinant proteins, to provide comparability of protein yields and detectable subunit clustering.

0 Q&A 259 Views Nov 5, 2025

This protocol describes the isolation and flow cytometric analysis of extracellular vesicles (EVs) derived from red blood cells, endothelial cells, and platelets in human peripheral blood. The protocol includes steps for preparing platelet-free plasma, fluorescent antibody staining, gating strategies, and technical controls. This protocol was developed within a study on EV release in snakebite-associated thrombotic microangiopathy; the protocol addresses challenges such as variable autofluorescence and heterogeneity in EV origin. It is flexible and can be adapted for alternative antibody panels targeting different cell populations or EV subtypes, including leukocyte-derived EVs.

0 Q&A 246 Views Nov 5, 2025

Cellular phenomena such as signal integration and transmission are based on the correct spatiotemporal organization of biomolecules within the cell. Therefore, the targeted manipulation of such processes requires tools that can precisely induce the localizations and interactions of the key players relevant to these processes with high temporal resolution. Chemically induced dimerization (CID) techniques offer a powerful means to manipulate protein function with high temporal resolution and subcellular specificity, enabling direct control over cellular behavior. Here, we present the detailed synthesis and application of dual SLIPT and dual SLIPTNVOC, which expand the SLIPT (self-localizing ligand-induced protein translocation) platform by incorporating a dual-ligand CID system. Dual SLIPT and dual SLIPTNVOC independently sort into the inner leaflet of the plasma membrane via a lipid-like anchoring motif, where they present the two headgroup moieties trimethoprim (TMP) and HaloTag ligand (HTL), which recruit and dimerize any two iK6eDHFR- and HOB-tagged proteins of interest (POIs). Dual-SLIPTNVOC furthermore enables this protein dimerization of POIs at the inner leaflet of the plasma membrane in a pre-determined order and light-controlled manner. In this protocol, we detail the synthetic strategy to access dual SLIPT and dual SLIPTNVOC, while also providing the underlying rationale for key design and synthetic decisions, with the aim of offering a streamlined, accessible, and broadly implementable methodology. In addition to the detailed synthesis, we present representative applications and typical experimental outcomes and recommend strategies for data analysis to support effective use of the system. Notably, dual SLIPT and dual SLIPTNVOC represent the first CID systems to emulate endogenous lipidation-driven membrane targeting, while retaining hallmark advantages of CID technology—the precision over POI identity, recruitment sequence, high spatiotemporal control, and “plug-and-play” flexibility.

0 Q&A 473 Views Nov 5, 2025

Three-dimensional (3D) human brain tissue models derived from induced pluripotent stem cells (iPSCs) have transformed the study of neural development and disease in vitro. While cerebral organoids offer high structural complexity, their large size often leads to necrotic core formation, limiting reproducibility and challenging the integration of microglia. Here, we present a detailed, reproducible protocol for generating multi-cell type 3D neurospheres that incorporate neurons, astrocytes, and optionally microglia, all derived from the same iPSCs. While neurons and astrocytes differentiate spontaneously from neural precursor cells, generated by dual SMAD-inhibition (blocking BMP and TGF-b signaling), microglia are generated in parallel and can infiltrate the mature neurosphere tissue after plating neurospheres into 48-well plates. The system supports a range of downstream applications, including functional confocal live imaging of GCaMP6f after adeno-associated virus (AAV) transduction of neurospheres or immunofluorescence staining after fixation. Our approach has been successfully implemented across multiple laboratories, demonstrating its robustness and translational potential for studying neuron–glia interactions and modeling neurodegenerative processes.

0 Q&A 229 Views Nov 5, 2025

Extracellular vesicles (EVs) have emerged as promising carriers for the targeted delivery of therapeutic proteins to specific cells. Previously, we demonstrated that genetically engineered EVs can be used for targeted protein delivery. This protocol details the generation of mannose receptor (CD206)-targeted EVs using a modular plasmid system optimized for production in HEK293T cells. Three plasmids enable customizable EV budding, cargo loading, and surface modification for targeting to antigen-presenting cells (APCs). EVs are isolated via differential centrifugation and chromatography, characterized using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA), and validated through functional uptake assays in primary human activated dendritic cells. Our approach combines flexibility in engineering required EVs with robust, reproducible isolation and characterization workflows. Its modularity allows easy adaptation to alternative targets or cargoes, which can be validated immediately through in vitro testing.

0 Q&A 215 Views Nov 5, 2025

Accurate labeling of excitatory postsynaptic sites remains a major challenge for high-resolution imaging due to the dense and sterically restricted environment of the postsynaptic density (PSD). Here, we present a protocol utilizing Sylites, 3 kDa synthetic peptide probes that bind with nanomolar affinity to key postsynaptic markers, PSD-95 and Gephyrin. eSylites (excitatory Sylites) specifically target the PDZ1 and PDZ2 domains of PSD-95, enabling precise and efficient labeling of excitatory postsynaptic density (ePSD). In contrast, iSylites (inhibitory Sylites) bind to the dimerizing E-domain of the Gephyrin C-terminus, allowing selective visualization of inhibitory postsynaptic density (iPSD). Their small size reduces linkage error and enhances accessibility compared to conventional antibodies, enabling clear separation of PSD-95 nanodomains in super-resolution microscopy. The protocol is compatible with co-labeling using standard antibodies and integrates seamlessly into multichannel immunocytochemistry workflows for primary neurons and brain tissue. This method enables robust, reproducible labeling of excitatory synapses with enhanced spatial resolution and can be readily adapted for expansion microscopy or live-cell applications.

Protocols in Past Issues
0 Q&A 710 Views Oct 20, 2025

Most membrane and secreted proteins are transported from the endoplasmic reticulum (ER) to the Golgi apparatus and subsequently directed to their final destinations in the cell. However, the mechanisms underlying transport and cargo sorting remain unclear. Recent advancements in optical microscopy, combined with synchronized cargo protein release methods, have enabled the direct observation of cargo protein transport. We developed a new optically synchronized cargo release method called retention using the dark state of LOV2 (RudLOV). This innovative technique offers three exceptional control capabilities: spatial, temporal, and quantitative control of cargo release. RudLOV uses illumination to trigger transport and detect cargo. Consequently, the selection of an appropriate laser and filter set for controlling the illumination and/or detection is crucial. The protocol presented here provides step-by-step guidelines for obtaining high-resolution live imaging data using RudLOV, thereby enabling researchers to investigate intracellular cargo transport with unprecedented precision and control.

0 Q&A 1014 Views Oct 20, 2025

Eukaryotic genomic DNA is packaged into chromatin, which plays a critical role in regulating gene expression by dynamically modulating its higher-order structure. While in vitro reconstitution approaches have offered valuable insights into chromatin organization, they often fail to fully capture the native structural context found within cells. To overcome this limitation, we present a protocol for isolating native chromatin fragments from human cells for cryo-electron microscopy (cryo-EM) analysis. In this method, chromatin from formaldehyde-crosslinked human HeLa S3 nuclei is digested with micrococcal nuclease (MNase) to generate mono- and poly-nucleosome fragments. These fragments are subsequently fractionated by sucrose-gradient ultracentrifugation and prepared for cryo-EM. The resulting chromatin fragments retain native-like nucleosome–nucleosome interactions, facilitating structural analyses of chromatin organization under near-physiological conditions.

0 Q&A 947 Views Oct 20, 2025

Adipose tissue macrophages (ATMs) critically influence obesity-induced inflammation and metabolic dysfunction. Recent studies identified distinct ATM subsets characterized by markers such as CD11c, CD9, and Trem2, associated with pro-inflammatory and metabolically activated states. This protocol outlines a detailed, reproducible methodology for isolating, characterizing, and sorting these ATM subsets from murine epididymal white adipose tissue (eWAT) using multicolor flow cytometry. Key steps include stromal vascular fraction (SVF) isolation, immunophenotyping, sequential gating strategies, and fluorescence-activated cell sorting (FACS) for downstream gene expression analysis. The protocol was validated in diet-induced obese (DIO) mice treated with the IRE1 RNase inhibitor STF-083010, demonstrating its utility for studying ATMs in the context of obesity and metabolic disease.

0 Q&A 474 Views Oct 20, 2025

Diabetes lacks concrete curative strategies due to diverse aetiologies and, therefore, represents the perfect candidate for cell replacement therapy, since it is caused by either an absolute (type 1 diabetes) or relative (type 2 diabetes) defect in the insulin-producing beta cells of the pancreas. Pancreatic alpha cells are a promising source for transdifferentiation into insulin-producing cells as they share a common developmental origin with beta cells and exhibit a certain degree of cellular plasticity. Furthermore, impairment of glucagon signaling in diabetes leads to a marked increase in alpha cell mass, raising the possibility that such alpha cell hyperplasia provides an increased supply of alpha cells for their transdifferentiation into new beta cells.

In this protocol, we used the modular epigenetic CRISPR/dCas9 toolbox for targeted DNA methylation (EpiCRISPR) and silencing of the Arx gene (Aristaless Related Homeobox, Arx), which is essential for the maintenance of alpha cell identity. Methylation-based silencing of Arx initiates the reprogramming of pancreatic alpha cells into insulin-producing cells. As a key novelty, this protocol provides a direct route for epigenetically induced transdifferentiation of mouse pancreatic alpha TC1-6 cells into insulin-producing cells and thereby confirms a proof of concept of reversible cellular epigenetic reprogramming in vitro. In addition, this streamlined workflow addresses the inherent challenges of transfecting clustered alpha TC1-6 cells by optimizing their dissociation into single-cell suspensions, thereby improving uptake and reproducibility.

In summary, this approach for cell transdifferentiation involves precise epigenetic editing of a lineage-specific marker gene, thereby enabling direct lineage conversion in a safe and versatile strategy to generate insulin-producing cells by epigenetic reprogramming. In contrast to approaches that rely on viral vectors or permanent genome editing, this method reduces the risk of off-target effects and immunogenic responses while ensuring reproducibility. The combination of efficiency and precision makes it a valuable tool to advance regenerative approaches for diabetes therapy and to explore the epigenetic regulation of cell identity.

0 Q&A 741 Views Oct 20, 2025

Translation is a key step in decoding the genetic information stored in DNA. Regulation of translation is an important step in gene expression control and is essential for healthy organismal development and behavior. Despite the importance of translation regulation, its impact and dynamics remain only partially understood. One reason is the lack of methods that enable the real-time visualization of translation in the context of multicellular organisms. To overcome this critical gap, microscopy-based methods that allow visualization of translation on single mRNAs in living cells and animals have been developed. A powerful approach is the SunTag system, which enables real-time imaging of nascent peptide synthesis with high spatial and temporal resolution. This protocol describes the implementation and use of the SunTag translation imaging system in the small round worm Caenorhabditis elegans. The protocol provides details on how to design, carry out, and interpret experiments to image translation dynamics of an mRNA of interest in a cell type of choice of living C. elegans. The ability to image translation live enables better understanding of translation and reveals the mechanisms underlying the dynamics of cell type–specific and subcellular localization of translation in development.

0 Q&A 1082 Views Oct 5, 2025

Intestinal organoids are generated from intestinal epithelial stem cells, forming 3D mini-guts that are often used as an in vitro model to evaluate and manipulate the regenerative capacities of intestinal epithelial stem cells. Plating 3D organoids on different substrates transforms organoids into 2D monolayers, which self-organize to form crypt-like regions (which contain stem cells and transit amplifying cells) and villus-like regions (which contain differentiated cells). This “open lumen” organization facilitates multiple biochemical and biomechanical studies that are otherwise complex in 3D organoids, such as drug applications to the cell’s apical side or precise control over substrate protein composition or substrate stiffness. Here, we describe a protocol to generate homogenous intestinal monolayers from single-cell intestinal organoid suspension, resulting in de novo crypt formation. Our protocol results in higher viability of intestinal cells, allowing successful monolayer formation.

0 Q&A 538 Views Oct 5, 2025

Inherited germline variants are now recognized as important contributors to hematologic myeloid malignancies, but their reliable detection depends on obtaining uncontaminated germline DNA. In solid tumors, peripheral blood remains free of tumor cells and therefore serves as a standard source for germline testing. In contrast, peripheral blood often contains neoplastic or clonally mutated cells in hematologic malignancies, making it impossible to distinguish somatic from germline variants. This unique challenge necessitates using an alternative, non-hematopoietic tissue source for accurate germline assessment in patients with hematologic myeloid malignancies. Cultured skin fibroblasts derived from punch biopsies have long been considered the gold standard for this purpose. Nevertheless, most existing protocols are optimized for research settings and lack detailed, patient-centric workflows for routine clinical use. Addressing this translational gap, we present a robust, enzyme-free protocol for culturing dermal fibroblasts from skin punch biopsies collected at the bedside during routine bone marrow procedures. The method details practical bedside collection, sterile transport, mechanical dissection without enzymatic digestion, plating strategy, culture expansion, and high-yield DNA isolation with validated purity. By integrating this standardized approach into routine hematopathology workflows, the protocol ensures reliable germline material with minimal patient discomfort and a turnaround time suitable for clinical diagnostics.

0 Q&A 854 Views Oct 5, 2025

Rapid and uniform labeling of plasma membrane proteins is essential for high-resolution imaging of dynamic membrane topologies and intercellular communication in live mammalian cells. Existing strategies for labeling live cell membranes, such as fluorescent fusion proteins, enzyme-mediated tags, metabolic bioorthogonal labeling, and lipophilic dyes, face trade-offs in the requirement of genetic manipulation, the presence of non-uniform labeling, the need for extensive preparation times, and limited choices of fluorophores. Here, we present a streamlined protocol that leverages N-hydroxysuccinimide (NHS)-ester chemistry to achieve rapid (≤5 min), covalent conjugation of synthetic small-molecule dyes to surface-exposed primary amines, enabling pan-membrane-protein labeling. This workflow covers dye stock preparation, labeling for suspension and adherent cells, multiplex live-cell imaging, fusion protein co-staining (including insulin-triggered receptor endocytosis), 3D membrane visualization, and in vivo assays for visualizing membrane-derived material transfers between donor and recipient cells using a lymphoma T-cell mouse model. This high-density labeling approach is compatible with various cell types across diverse imaging platforms. Its speed, versatility, and stability make it a broadly applicable tool for studying plasma membrane dynamics and intercellular membrane trafficking.

0 Q&A 497 Views Oct 5, 2025

Here, we present a protocol for implementing the fluorogen-activating protein FAST (fluorescence-activating and absorption-shifting tag) in fluorescence lifetime imaging microscopy (FLIM), which allows separating fluorescent species in the same spectral channel based on fluorescence lifetime properties. Previous studies have demonstrated FLIM multiplexing using various combinations of synthetic probes, fluorescent proteins, or self-labeling tags. In this protocol, we utilize engineered FAST point mutation variants that bind fluorogen HBR-2,5-DM. The designed probes possess nearly identical, compact protein sizes (14 kDa), and the resulting protein–fluorogen complexes demonstrate comparable steady-state optical properties and exhibit distinct fluorescence lifetimes, displaying monoexponential fluorescence decay kinetics. When FAST variants are expressed with localization signals, these properties facilitate robust signal separation in regions with co-localized or spatially overlapping labels (nucleus and cytoskeleton in this protocol) in live mammalian cells. This method can be applied to separate other overlapping cellular compartments, such as the nucleus and Golgi apparatus, or mitochondria and cytoskeleton.

0 Q&A 825 Views Oct 5, 2025

High-content analysis (HCA) is a powerful image-based approach for phenotypic profiling and drug discovery, enabling the extraction of multiparametric data from individual cells. Traditional HCA protocols often rely on fixed-cell imaging, with assays like cell painting widely adopted as standard. While these methods provide rich morphological information, the integration of live-cell imaging expands analytical capabilities by enabling the study of dynamic biological processes and real-time cellular responses. This protocol presents a simple, cost-effective, and scalable method for live-cell HCA using acridine orange (AO), a metachromatic fluorescent dye that highlights cellular organization by staining nucleic acids and acidic compartments. The assay provides visualization of distinct subcellular structures, including nuclei and cytoplasmic organelles, using a two-channel fluorescence readout. Compatible with high-throughput microscopy and computational analysis, the method supports diverse applications such as phenotypic screening, cytotoxicity assessment, and morphological profiling. By preserving cell viability and enabling dynamic, real-time measurements, this live-cell imaging approach complements existing fixed-cell assays and offers a versatile platform for uncovering complex cellular phenotypes.




We use cookies to improve your user experience on this site. By using our website, you agree to the storage of cookies on your computer.
点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载