In conclusion, the creation of new techniques and tools to enable the study of fundamental EV biology holds significant value for the advancement of the field. Approaches to monitor EV production and release are frequently based on either antibody-based flow cytometry assays or genetically encoded fluorescent proteins. VH298 datasheet In prior work, we engineered artificially barcoded exosomal microRNAs (bEXOmiRs) to serve as high-throughput reporters of extracellular vesicle release. The initial phase of this protocol meticulously outlines the essential steps and factors to consider in the development and replication of bEXOmiRs. We now proceed to describe the analysis of bEXOmiR expression and abundance in cells, as well as in isolated extracellular vesicles.
The transfer of nucleic acids, proteins, and lipid molecules between cells relies on the function of extracellular vesicles (EVs). Genetic, physiological, and pathological modifications in the recipient cell can arise from biomolecular cargo carried within extracellular vesicles. Electric vehicles' inbuilt capacity enables the transportation of pertinent cargo to a defined cell or organ. Extracellular vesicles (EVs), possessing the remarkable ability to permeate the blood-brain barrier (BBB), are effectively employed as delivery vehicles for therapeutic drugs and substantial macromolecules to hard-to-reach organs such as the brain. Therefore, laboratory techniques and protocols, focusing on the modification of EVs, are presented in this chapter to support neuronal research.
Exosomes, small extracellular vesicles, measuring 40 to 150 nanometers in diameter, are discharged by nearly all cell types and function in dynamic intercellular and interorgan communication processes. Vesicles secreted by source cells transport diverse biologically active components, encompassing microRNAs (miRNAs) and proteins, consequently altering the molecular functionalities of target cells in distant tissues. Accordingly, exosomes are integral to controlling critical functions performed by microenvironments inside tissues. The exact methodologies by which exosomes bind to and migrate to particular organs remained largely unclear. Over recent years, the significant family of cell-adhesion molecules, integrins, have been discovered to be fundamental in directing the targeting of exosomes to specific tissues, since integrins manage the tissue-specific homing of cells. It is imperative to experimentally determine how integrins influence the tissue-specific targeting of exosomes. This chapter outlines a protocol for investigating the integrin-mediated targeting of exosomes, considering both in vitro and in vivo experimental environments. VH298 datasheet The study of integrin 7 is our primary focus, as its function in lymphocyte gut-specific homing has been well-characterized.
Within the EV research community, the study of the molecular pathways governing extracellular vesicle uptake by a target cell is a significant focus. This reflects the critical function of EVs in mediating intercellular communication, which is essential for tissue homeostasis or for impacting disease progression, like cancer and Alzheimer's. In light of the relatively young age of the EV sector, the standardization of methods for even basic procedures like isolation and characterization is an ongoing process and a subject of debate. The study of electric vehicle adoption similarly reveals that current strategies are fundamentally hampered. Improving the sensitivity and reliability of the assays, and/or separating surface EV binding from uptake events, should be a focus of new approaches. Two supplementary strategies for gauging and quantifying EV adoption are presented here. We believe these methods will address some limitations of existing techniques. Employing a mEGFP-Tspn-Rluc construct allows for the sorting of these two reporters into EVs. The capacity to measure EV uptake through bioluminescence signaling boosts sensitivity, allows for the determination of EV binding versus cellular internalization, and allows for kinetics analysis in living cells, aligning with the requirements of high-throughput screening. As a second approach, a flow cytometry assay is developed, relying on maleimide-fluorophore conjugate-labeled EVs. This chemical compound binds covalently to proteins with sulfhydryl residues, offering a promising alternative to lipid-based dyes. The method is compatible with flow cytometry sorting of cell populations that have incorporated the labeled EVs.
Exosomes, minuscule sacs that are released by each and every type of cell, are hypothesized to serve as a promising and natural pathway for the exchange of information between cells. The delivery of exosomes' internal contents to cells in close proximity or at a distance may contribute to mediating intercellular communication. This newly discovered exosome cargo transfer capability has sparked the development of a new therapeutic strategy, and exosomes are being examined as vehicles for delivering cargo, especially nanoparticles (NPs). This report elucidates the process of NP encapsulation, achieved by incubating cells with NPs, along with the subsequent methods used to identify the cargo and prevent detrimental changes in the loaded exosomes.
The development and progression of a tumor, including resistance to antiangiogenesis therapies (AATs), is subject to substantial regulation by exosomes. Exosomes originate from a dual source: tumor cells and the encompassing endothelial cells (ECs). The methods employed to analyze cargo transfer between tumor cells and endothelial cells (ECs), using a novel four-compartment co-culture system, are detailed. Also detailed is the evaluation of how tumor cells affect the angiogenic ability of ECs through the use of Transwell co-culture.
Biomacromolecular separation from human plasma, achieved using immunoaffinity chromatography (IAC) with antibodies on polymeric monolithic disk columns, is followed by further fractionation into specific subpopulations, including small dense low-density lipoproteins, exomeres, and exosomes, by asymmetrical flow field-flow fractionation (AsFlFFF or AF4). The process of isolating and fractionating subpopulations of extracellular vesicles, free from lipoproteins, is presented here, utilizing the on-line coupled IAC-AsFlFFF approach. Using the developed methodology, fast, reliable, and reproducible automated isolation and fractionation of challenging biomacromolecules from human plasma can be achieved, resulting in high purity and high yields of subpopulations.
For the successful development of a therapeutic product derived from extracellular vesicles (EVs), reliable and scalable purification protocols for clinical-grade EVs must be incorporated. Commonly utilized methods of isolation, encompassing ultracentrifugation, density gradient centrifugation, size exclusion chromatography, and polymer-based precipitation, exhibited shortcomings in terms of yield effectiveness, vesicle purity, and sample volume limitations. For the scalable production, concentration, and isolation of EVs, a GMP-compliant method employing tangential flow filtration (TFF) was created. Using this purification technique, we isolated extracellular vesicles (EVs) from the conditioned medium (CM) of cardiac stromal cells, specifically cardiac progenitor cells (CPCs), known for their potential therapeutic applications in managing heart failure. Exosome vesicle (EV) isolation using tangential flow filtration (TFF) from conditioned media exhibited a consistent particle recovery, approximately 10^13 per milliliter, focusing on enriching the 120-140 nanometer size range of exosomes. Major protein-complex contaminant levels in EV preparations were reduced by a substantial 97%, resulting in no change to their biological activity. The protocol details the assessment of EV identity and purity, and subsequent procedures for applications, including functional potency testing and quality control procedures. A versatile protocol, easily adaptable to a variety of cell sources, is exemplified by large-scale GMP-grade electric vehicle manufacturing, applicable to a wide range of therapeutic areas.
The discharge of extracellular vesicles (EVs), along with their constituent components, is responsive to a range of clinical circumstances. Extracellular vesicles (EVs), participating in intercellular communication, are hypothesized to mirror the pathophysiology of the cells, tissues, organs or the system they interface with. Urinary EVs effectively demonstrate the pathophysiological characteristics of renal diseases, acting as an auxiliary source of potential biomarkers accessible without invasive procedures. VH298 datasheet The primary focus on the cargo in electric vehicles has been proteins and nucleic acids, with a recent addition of metabolites to that interest. As a reflection of processes occurring within living organisms, the genome, transcriptome, and proteome's downstream modifications are observed as changes in metabolites. Nuclear magnetic resonance (NMR) and liquid chromatography-mass spectrometry (LC-MS/MS) are commonly utilized in their research. NMR spectroscopy stands as a reliable and nondestructive method, and we present here the methodological protocols for urinary exosome metabolomic analysis using NMR. Besides describing the workflow for a targeted LC-MS/MS analysis, we discuss its expansion to untargeted studies.
The separation of extracellular vesicles (EVs) from conditioned cell culture media has been a difficult issue. Large-scale procurement of pristine, unaltered EVs presents a significant challenge. Differential centrifugation, ultracentrifugation, size exclusion chromatography, polyethylene glycol (PEG) precipitation, filtration, and affinity-based purification, amongst other widely employed techniques, exhibit varying degrees of benefit and drawback. A multi-step purification protocol, utilizing tangential-flow filtration (TFF), is presented, which combines filtration, PEG precipitation, and Capto Core 700 multimodal chromatography (MMC) to yield highly pure EVs from substantial quantities of cell culture conditioned medium. Implementing the TFF stage before PEG precipitation minimizes protein buildup, potentially preventing their aggregation and co-purification with extracellular vesicles.