1
|
Kotak D, Attar E, Dalal B, Shankarkumar A, Devarajan P. Quantitative Analysis of Salmon Calcitonin Hydroxyapatite Nanoparticle Permeation to substantiate Non-Invasive Bone Targeting via Sublingual Delivery. AAPS PharmSciTech 2025; 26:88. [PMID: 40102334 DOI: 10.1208/s12249-025-03068-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
We earlier reported comparable efficacy in bone parameters of sublingually administered salmon calcitonin hydroxyapatite nanoparticles (SCT-HAP-NPs) compared to the subcutaneous injection, in the ovariectomy rat model, despite a bioavailability of barely ~ 15%. We ascribed this intriguing finding to targeted bone delivery, facilitated by translocation of significant quantity of intact NP into systemic circulation. In the present study we track the translocation of FITC-SCT-HAP-NPs (~ 100 nm) across porcine sublingual mucosa using the Franz diffusion cell to validate our hypothesis. Confocal Laser Scanning microscopy (CLSM) established that SCT-HAP-NPs permeated into the deeper layers of sublingual porcine mucosal tissue. We confirmed the nanoparticles were present in the receptor medium of the Franz diffusion cell by DLS and TEM. We also demonstrate for the first time quantification of the NPs (%) translocated across the porcine mucosa, using the Amnis Image StreamX Mk II imaging flow cytometer. Computation revealed transport of ~ 60% of the FITC-SCT-HAP-NPs across mucosa in 2 h, substantiated that high NP concentrations could reach systemic circulation. Such high NP concentration in systemic circulation coupled with the small size (~ 100 nm) and the high bone affinity of HAP, validate our hypothesis of targeted bone delivery following sublingual administration. Furthermore, quantification of translocated NPs, which we demonstrate for the first time, would permit rational development of optimal targeted nanoparticulate carriers for delivery by noninvasive routes.
Collapse
Affiliation(s)
- Darsheen Kotak
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga (E), Mumbai, 400019, Maharashtra, India
| | - Esha Attar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga (E), Mumbai, 400019, Maharashtra, India
| | - Bhavik Dalal
- Department of Transfusion Transmitted Disease, ICMR-National Institute of Immunohematology, KEM Hospital Campus, Parel, Mumbai, 400012, Maharashtra, India
| | - Aruna Shankarkumar
- Department of Transfusion Transmitted Disease, ICMR-National Institute of Immunohematology, KEM Hospital Campus, Parel, Mumbai, 400012, Maharashtra, India
| | - Padma Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga (E), Mumbai, 400019, Maharashtra, India.
| |
Collapse
|
2
|
Waliullah ASM, Qiu K, Dziegielewska B, Tran ML, Nguyen NN, Wang L, Pan A, Segovia N, Umarino S, Zhang J, Nguyen TM, Craig J, Tenen DG, Trinh BQ. An integrated DNA interactome and transcriptome profiling reveals a PU.1/enhancer RNA-mediated Feed-forward Regulatory Loop Regulating monocyte/macrophage development and innate immune functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.638695. [PMID: 40027734 PMCID: PMC11870581 DOI: 10.1101/2025.02.19.638695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
High expression of the myeloid master ETS transcription factor PU.1 drives the development of monocyte/macrophage (Mono/MΦ), a crucial cellular component of the innate immune system. Disruptions in normal expression patterns of PU.1 are linked to a variety myeloid malignancy and immune diseases. It is evidenced that PU.1 binds to and modulates enhancers of several myeloid genes. While noncoding RNAs transcribed from noncoding genes at the enhancers are increasingly reported to be involved in enhancer regulation, the crosstalk between PU.1 and noncoding RNAs in enhancer-mediated myeloid gene regulation in Mono/MΦ differentiation and immune response has not been systematically investigated. In this study, we interrogated the PU.1-mediated transcriptome and cistrome with our comprehensive collection of putative and verified enhancers. Among a repertoire of noncoding genes present at PU.1-bound enhancers, we discovered that PU.1 acts as a potent transcription factor inducer of the noncoding RNA LOUP , which we previously identified as an RNA inducer of PU.1. The genomic region within the LOUP locus occupied by PU.1 is characterized by the epigenetic features of a myeloid-specific super-enhancer. Targeted disruption of the PU.1-binding motifs resulted in the downregulation of LOUP promoter activity. Depletion of LOUP reduced the expression of Mono/MΦ cell markers as well as the transcriptional program associated with Mono/MΦ differentiation Mono/MΦ innate defense mechanisms, including phagocytosis, antimicrobial activity, and chemoattractant cytokine production. LOUP induces Mono/MΦ phagocytic activities. Collectively, our findings indicate that PU.1 and enhancer RNA LOUP are biomolecular components of an unidentified feed-forward loop that promotes their mutual expression, contributing to Mono/MΦ differentiation and innate immune functions. The identification of the PU.1/ LOUP regulatory circuit provides valuable insights into the mechanisms underlying cell-type and gene-specific enhancer activity and Mono/MΦ biology, as well as significant implications for advancing our understanding of immune diseases and myeloid malignancies.
Collapse
|
3
|
He W, Yan L, Hu D, Hao J, Liou Y, Luo G. Neutrophil heterogeneity and plasticity: unveiling the multifaceted roles in health and disease. MedComm (Beijing) 2025; 6:e70063. [PMID: 39845896 PMCID: PMC11751288 DOI: 10.1002/mco2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Neutrophils, the most abundant circulating leukocytes, have long been recognized as key players in innate immunity and inflammation. However, recent discoveries unveil their remarkable heterogeneity and plasticity, challenging the traditional view of neutrophils as a homogeneous population with a limited functional repertoire. Advances in single-cell technologies and functional assays have revealed distinct neutrophil subsets with diverse phenotypes and functions and their ability to adapt to microenvironmental cues. This review provides a comprehensive overview of the multidimensional landscape of neutrophil heterogeneity, discussing the various axes along which diversity manifests, including maturation state, density, surface marker expression, and functional polarization. We highlight the molecular mechanisms underpinning neutrophil plasticity, focusing on the complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications that shape neutrophil responses. Furthermore, we explore the implications of neutrophil heterogeneity and plasticity in physiological processes and pathological conditions, including host defense, inflammation, tissue repair, and cancer. By integrating insights from cutting-edge research, this review aims to provide a framework for understanding the multifaceted roles of neutrophils and their potential as therapeutic targets in a wide range of diseases.
Collapse
Affiliation(s)
- Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Dongxue Hu
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| |
Collapse
|
4
|
Di S, Huang Y, Qiao W, Zhang X, Wang Y, Zhang M, Fu J, Zhao J, Chen L. Advances in the isolation and characterization of milk-derived extracellular vesicles and their functions. Front Nutr 2024; 11:1512939. [PMID: 39742102 PMCID: PMC11688093 DOI: 10.3389/fnut.2024.1512939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Milk-derived extracellular vesicles (EVs) have various functions, including immune regulation and promoting intestinal development. These EVs have substantial potential for application in infant formula and functional foods development. In addition, numerous studies have shown that milk-derived EVs carry proteins, lipids, and nucleic acids away from their parental cells, acting as messengers between cells. Moreover, structural integrity and biological viability are necessary prerequisites for the functional and omics studies of milk-derived EVs. Therefore, selecting appropriate methods for isolating and characterizing milk-derived EVs is essential for subsequent studies. Accordingly, this review summarizes the isolation and characterization methods for milk-derived EVs and their biological functions and roles. Furthermore, it discusses the comprehensive application of isolation methods, providing a reference for research on and development of milk-derived EVs.
Collapse
Affiliation(s)
- Shujuan Di
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yibo Huang
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Xiaomei Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yaling Wang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Minghui Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Jieyu Fu
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lijun Chen
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| |
Collapse
|
5
|
Visser C, Rivieccio F, Krüger T, Schmidt F, Cseresnyés Z, Rohde M, Figge MT, Kniemeyer O, Blango MG, Brakhage AA. Tracking the uptake of labelled host-derived extracellular vesicles by the human fungal pathogen Aspergillus fumigatus. MICROLIFE 2024; 5:uqae022. [PMID: 39660046 PMCID: PMC11631206 DOI: 10.1093/femsml/uqae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
Extracellular vesicles (EVs) have gained attention as facilitators of intercellular as well as interkingdom communication during host-microbe interactions. Recently we showed that upon infection, host polymorphonuclear leukocytes produce antifungal EVs targeting the clinically important fungal pathogen Aspergillus fumigatus; however, the small size of EVs (<1 µm) complicates their functional analysis. Here, we employed a more tractable, reporter-based system to label host alveolar epithelial cell-derived EVs and enable their visualization during in vitro A. fumigatus interaction. Fusion of EV marker proteins (CD63, CD9, and CD81) with a Nanoluciferase (NLuc) and a green fluorescent protein (GFP) facilitated their relative quantification by luminescence and visualization by a fluorescence signal. The use of an NLuc fused with a GFP is advantageous as it allows for quantification and visualization of EVs simultaneously without additional external manipulation and to distinguish subpopulations of EVs. Using this system, visualization and tracking of EVs was possible using confocal laser scanning microscopy and advanced imaging analysis. These experiments revealed the propensity of host cell-derived EVs to associate with the fungal cell wall and ultimately colocalize with the cell membrane of A. fumigatus hyphae in large numbers. In conclusion, we have created a series of tools to better define the complex interplay of host-derived EVs with microbial pathogens.
Collapse
Affiliation(s)
- Corissa Visser
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Flora Rivieccio
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Franziska Schmidt
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Zoltán Cseresnyés
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
- Excellence Cluster Balance of the Microverse, Friedrich Schiller University, 07743 Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Matthew G Blango
- Junior Research Group RNA Biology of Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
- Excellence Cluster Balance of the Microverse, Friedrich Schiller University, 07743 Jena, Germany
| |
Collapse
|
6
|
Jhaveri JR, Khare P, Paul Pinky P, Kamte YS, Chandwani MN, Milosevic J, Abraham N, Sun M, Stolz DB, Dave KM, Zheng SY, O'Donnell L, Manickam DS. Low pinocytic brain endothelial cells primarily utilize membrane fusion to internalize extracellular vesicles. Eur J Pharm Biopharm 2024; 204:114500. [PMID: 39303949 DOI: 10.1016/j.ejpb.2024.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Extracellular vesicles (EVs) are an emerging class of drug carriers and are primarily reported to be internalized into recipient cells via a combination of endocytic routes such as clathrin-mediated, caveolae-mediated and macropinocytosis pathways. In this work, (1) we investigated potential effects of homotypic vs. heterotypic interactions by studying the cellular uptake of homologous EVs (EV donor cells and recipient cells of the same type) vs. heterologous EVs (EV donor cells and recipient cells of different types) and (2) determined the route of EV internalization into low pinocytic/hard-to-deliver cell models such as brain endothelial cells (BECs). Homotypic interactions led to a greater extent of uptake into the recipient BECs compared to heterotypic interactions. However, we did not see a complete reduction in EV uptake into recipient BECs when endocytic pathways were blocked using pharmacological inhibitors and our findings from a R18-based fusion assay suggest that EVs primarily use membrane fusion to enter low-pinocytic recipient BECs instead of relying on endocytosis. Lipophilic PKH67 dye-labeled EVs but not intravesicular esterase-activated calcein ester-labeled EVs severely reduced particle uptake into BECs while phagocytic macrophages internalized EVs labeled with both dyes to comparable extents. Our results also highlight the importance of carefully choosing labeling dye chemistry to study EV uptake, especially in the case of low pinocytic cells such as BECs.
Collapse
Affiliation(s)
- Jhanvi R Jhaveri
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Paromita Paul Pinky
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Yashika S Kamte
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Manisha N Chandwani
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States; Captis Diagnostics Inc., Pittsburgh, PA, United States
| | - Nevil Abraham
- Unified Flow Core, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ming Sun
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Lauren O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.
| |
Collapse
|
7
|
Woud WW, Pugsley HR, Bettin BA, Varga Z, van der Pol E. Size and fluorescence calibrated imaging flow cytometry: From arbitrary to standard units. Cytometry A 2024; 105:752-762. [PMID: 39238272 DOI: 10.1002/cyto.a.24895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
Imaging flow cytometry (IFCM) is a technique that can detect, size, and phenotype extracellular vesicles (EVs) at high throughput (thousands/minute) in complex biofluids without prior EV isolation. However, the generated signals are expressed in arbitrary units, which hinders data interpretation and comparison of measurement results between instruments and institutes. While fluorescence calibration can be readily achieved, calibration of side scatter (SSC) signals presents an ongoing challenge for IFCM. Here, we present an approach to relate the SSC signals to particle size for IFCM, and perform a comparability study between three different IFCMs using a plasma EV test sample (PEVTES). SSC signals for different sizes of polystyrene (PS) and hollow organosilica beads (HOBs) were acquired with a 405 nm 120 mW laser without a notch filter before detection. Mie theory was applied to relate scatter signals to particle size. Fluorescence calibration was accomplished with 2 μm phycoerythrin (PE) and allophycocyanin (APC) MESF beads. Size and fluorescence calibration was performed for three IFCMs in two laboratories. CD235a-PE and CD61-APC stained PEVTES were used as EV-containing samples. EV concentrations were compared between instruments within a size range of 100-1000 nm and a fluorescence intensity range of 3-10,000 MESF. 81 nm PS beads could be readily discerned from background based on their SSC signals. Fitting of the obtained PS bead SSC signals with Mie theory resulted in a coefficient of determination >0.99 between theory and data for all three IFCMs. 216 nm HOBs were detected with all instruments, and confirmed the sensitivity to detect EVs by SSC. The lower limit of detection regarding EV-size for this study was determined to be ~100 nm for all instruments. Size and fluorescence calibration of IFCM data increased cross-instrument data comparability with the coefficient of variation decreasing from 33% to 21%. Here we demonstrate - for the first time - scatter calibration of an IFCM using the 405 nm laser. The quality of the scatter-to-diameter relation and scatter sensitivity of the IFCMs are similar to the most sensitive commercially available flow cytometers. This development will support the reliability of EV research with IFCM by providing robust standardization and reproducibility, which are pre-requisites for understanding the biological significance of EVs.
Collapse
Affiliation(s)
- Wouter W Woud
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Haley R Pugsley
- Application Cytometry, Cytek Biosciences, Inc, Seattle, Washington, USA
| | - Britta A Bettin
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Chemistry, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
| | - Zoltán Varga
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Edwin van der Pol
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Chemistry, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Padinharayil H, George A. Small extracellular vesicles: Multi-functional aspects in non-small cell lung carcinoma. Crit Rev Oncol Hematol 2024; 198:104341. [PMID: 38575042 DOI: 10.1016/j.critrevonc.2024.104341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Extracellular vesicles (EVs) impact normal and pathological cellular signaling through bidirectional trafficking. Exosomes, a subset of EVs possess biomolecules including proteins, lipids, DNA fragments and various RNA species reflecting a speculum of their parent cells. The involvement of exosomes in bidirectional communication and their biological constituents substantiate its role in regulating both physiology and pathology, including multiple cancers. Non-small cell lung cancer (NSCLC) is the most common lung cancers (85%) with high incidence, mortality and reduced overall survival. Lack of efficient early diagnostic and therapeutic tools hurdles the management of NSCLC. Interestingly, the exosomes from body fluids similarity with parent cells or tissue offers a potential future multicomponent tool for the early diagnosis of NSCLC. The structural twinning of exosomes with a cell/tissue and the competitive tumor derived exosomes in tumor microenvironment (TME) promotes the unpinning horizons of exosomes as a drug delivery, vaccine, and therapeutic agent. Exosomes in clinical point of view assist to trace: acquired resistance caused by various therapeutic agents, early diagnosis, progression, and surveillance. In an integrated approach, EV biomarkers offer potential cutting-edge techniques for the detection and diagnosis of cancer, though the purification, characterization, and biomarker identification processes for the translational research regarding EVs need further optimization.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India.
| |
Collapse
|
9
|
Bhavsar D, Raguraman R, Kim D, Ren X, Munshi A, Moore K, Sikavitsas V, Ramesh R. Exosomes in diagnostic and therapeutic applications of ovarian cancer. J Ovarian Res 2024; 17:113. [PMID: 38796525 PMCID: PMC11127348 DOI: 10.1186/s13048-024-01417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/16/2024] [Indexed: 05/28/2024] Open
Abstract
Ovarian cancer accounts for more deaths than any other female reproductive tract cancer. The major reasons for the high mortality rates include delayed diagnoses and drug resistance. Hence, improved diagnostic and therapeutic options for ovarian cancer are a pressing need. Extracellular vesicles (EVs), that include exosomes provide hope in both diagnostic and therapeutic aspects. They are natural lipid nanovesicles secreted by all cell types and carry molecules that reflect the status of the parent cell. This facilitates their potential use as biomarkers for an early diagnosis. Additionally, EVs can be loaded with exogenous cargo, and have features such as high stability and favorable pharmacokinetic properties. This makes them ideal for tumor-targeted delivery of biological moieties. The International Society of Extracellular Vesicles (ISEV) based on the Minimal Information for Studies on Extracellular Vesicles (MISEV) recommends the usage of the term "small extracellular vesicles (sEVs)" that includes exosomes for particles that are 30-200 nm in size. However, majority of the studies reported in the literature and relevant to this review have used the term "exosomes". Therefore, this review will use the term "exosomes" interchangeably with sEVs for consistency with the literature and avoid confusion to the readers. This review, initially summarizes the different isolation and detection techniques developed to study ovarian cancer-derived exosomes and the potential use of these exosomes as biomarkers for the early diagnosis of this devastating disease. It addresses the role of exosome contents in the pathogenesis of ovarian cancer, discusses strategies to limit exosome-mediated ovarian cancer progression, and provides options to use exosomes for tumor-targeted therapy in ovarian cancer. Finally, it states future research directions and recommends essential research needed to successfully transition exosomes from the laboratory to the gynecologic-oncology clinic.
Collapse
Affiliation(s)
- Dhaval Bhavsar
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE, 10th Street, Oklahoma City, OK, 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
| | - Rajeswari Raguraman
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE, 10th Street, Oklahoma City, OK, 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N, Stonewall Ave, Oklahoma City, OK, 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
| | - Xiaoyu Ren
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N, Stonewall Ave, Oklahoma City, OK, 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, 975 NE, 10th Street, Oklahoma City, OK, 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
| | - Kathleen Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
| | - Vassilios Sikavitsas
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA
- Department of Chemical, Biological and Materials Engineering, Oklahoma University, Norman, OK, 73019, USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE, 10th Street, Oklahoma City, OK, 73104, USA.
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 800 NE, 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
10
|
Dimitriadis S, Dova L, Kotsianidis I, Hatzimichael E, Kapsali E, Markopoulos GS. Imaging Flow Cytometry: Development, Present Applications, and Future Challenges. Methods Protoc 2024; 7:28. [PMID: 38668136 PMCID: PMC11054958 DOI: 10.3390/mps7020028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/29/2024] Open
Abstract
Imaging flow cytometry (ImFC) represents a significant technological advancement in the field of cytometry, effectively merging the high-throughput capabilities of flow analysis with the detailed imaging characteristics of microscopy. In our comprehensive review, we adopt a historical perspective to chart the development of ImFC, highlighting its origins and current state of the art and forecasting potential future advancements. The genesis of ImFC stemmed from merging the hydraulic system of a flow cytometer with advanced camera technology. This synergistic coupling facilitates the morphological analysis of cell populations at a high-throughput scale, effectively evolving the landscape of cytometry. Nevertheless, ImFC's implementation has encountered hurdles, particularly in developing software capable of managing its sophisticated data acquisition and analysis needs. The scale and complexity of the data generated by ImFC necessitate the creation of novel analytical tools that can effectively manage and interpret these data, thus allowing us to unlock the full potential of ImFC. Notably, artificial intelligence (AI) algorithms have begun to be applied to ImFC, offering promise for enhancing its analytical capabilities. The adaptability and learning capacity of AI may prove to be essential in knowledge mining from the high-dimensional data produced by ImFC, potentially enabling more accurate analyses. Looking forward, we project that ImFC may become an indispensable tool, not only in research laboratories, but also in clinical settings. Given the unique combination of high-throughput cytometry and detailed imaging offered by ImFC, we foresee a critical role for this technology in the next generation of scientific research and diagnostics. As such, we encourage both current and future scientists to consider the integration of ImFC as an addition to their research toolkit and clinical diagnostic routine.
Collapse
Affiliation(s)
- Savvas Dimitriadis
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
| | - Lefkothea Dova
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
| | - Ioannis Kotsianidis
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.H.); (E.K.)
| | - Eleni Kapsali
- Department of Hematology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.H.); (E.K.)
| | - Georgios S. Markopoulos
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
- Department of Surgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
11
|
Fischer Weinberger R, Bachmaier S, Ober V, Githure GB, Dandugudumula R, Phan IQ, Almoznino M, Polatoglou E, Tsigankov P, Nitzan Koren R, Myler PJ, Boshart M, Zilberstein D. A divergent protein kinase A regulatory subunit essential for morphogenesis of the human pathogen Leishmania. PLoS Pathog 2024; 20:e1012073. [PMID: 38551993 PMCID: PMC11006142 DOI: 10.1371/journal.ppat.1012073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/10/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Parasitic protozoa of the genus Leishmania cycle between the phagolysosome of mammalian macrophages, where they reside as rounded intracellular amastigotes, and the midgut of female sand flies, which they colonize as elongated extracellular promastigotes. Previous studies indicated that protein kinase A (PKA) plays an important role in the initial steps of promastigote differentiation into amastigotes. Here, we describe a novel regulatory subunit of PKA (which we have named PKAR3) that is unique to Leishmania and most (but not all) other Kinetoplastidae. PKAR3 is localized to subpellicular microtubules (SPMT) in the cell cortex, where it recruits a specific catalytic subunit (PKAC3). Promastigotes of pkar3 or pkac3 null mutants lose their elongated shape and become rounded but remain flagellated. Truncation of an N-terminal formin homology (FH)-like domain of PKAR3 results in its detachment from the SPMT, also leading to rounded promastigotes. Thus, the tethering of PKAC3 via PKAR3 at the cell cortex is essential for maintenance of the elongated shape of promastigotes. This role of PKAR3 is reminiscent of PKARIβ and PKARIIβ binding to microtubules of mammalian neurons, which is essential for the elongation of dendrites and axons, respectively. Interestingly, PKAR3 binds nucleoside analogs, but not cAMP, with a high affinity similar to the PKAR1 isoform of Trypanosoma. We propose that these early-diverged protists have re-purposed PKA for a novel signaling pathway that spatiotemporally controls microtubule remodeling and cell shape.
Collapse
Affiliation(s)
| | - Sabine Bachmaier
- Faculty of Biology, Genetics, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Veronica Ober
- Faculty of Biology, Genetics, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - George B. Githure
- Faculty of Biology, Genetics, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Ramu Dandugudumula
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Isabelle Q. Phan
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Michal Almoznino
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Eleni Polatoglou
- Faculty of Biology, Genetics, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Polina Tsigankov
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Roni Nitzan Koren
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Peter J. Myler
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pediatrics, Department of Biomedical Informatics & Medical Education, and Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Michael Boshart
- Faculty of Biology, Genetics, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Dan Zilberstein
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
12
|
Jyoti TP, Chandel S, Singh R. Flow cytometry: Aspects and application in plant and biological science. JOURNAL OF BIOPHOTONICS 2024; 17:e202300423. [PMID: 38010848 DOI: 10.1002/jbio.202300423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/28/2023] [Indexed: 11/29/2023]
Abstract
Flow cytometry is a potent method that enables the quick and concurrent investigation of several characteristics of single cells in solution. Photodiodes or photomultiplier tubes are employed to detect the dispersed and fluorescent light signals that are produced by the laser beam as it passes through the cells. Photodetectors transform the light signals produced by the laser into electrical impulses. A computer then analyses these electrical impulses to identify and measure the various cell populations depending on their fluorescence or light scattering characteristics. Based on their fluorescence or light scattering properties, cell populations can be examined and/or isolated. This review covers the basic principle, components, working and specific biological applications of flow cytometry, including studies on plant, cell and molecular biology and methods employed for data processing and interpretation as well as the potential future relevance of this methodology in light of retrospective analysis and recent advancements in flow cytometry.
Collapse
Affiliation(s)
- Thakur Prava Jyoti
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab, India
| | - Shivani Chandel
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab, India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
13
|
Tsoneva DK, Ivanov MN, Vinciguerra M. Liquid Liver Biopsy for Disease Diagnosis and Prognosis. J Clin Transl Hepatol 2023; 11:1520-1541. [PMID: 38161500 PMCID: PMC10752811 DOI: 10.14218/jcth.2023.00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/24/2023] [Accepted: 06/07/2023] [Indexed: 01/03/2024] Open
Abstract
Liver diseases are a major burden worldwide, the scope of which is expected to further grow in the upcoming years. Clinically relevant liver dysfunction-related blood markers such as alanine aminotransferase and aspartate aminotransferase have limited accuracy. Nowadays, liver biopsy remains the gold standard for several liver-related pathologies, posing a risk of complication due to its invasive nature. Liquid biopsy is a minimally invasive approach, which has shown substantial potential in the diagnosis, prognosis, and monitoring of liver diseases by detecting disease-associated particles such as proteins and RNA molecules in biological fluids. Histones are the core components of the nucleosomes, regulating essential cellular processes, including gene expression and DNA repair. Following cell death or activation of immune cells, histones are released in the extracellular space and can be detected in circulation. Histones are stable in circulation, have a long half-life, and retain their post-translational modifications. Here, we provide an overview of the current research on histone-mediated liquid biopsy methods for liver diseases, with a focus on the most common detection methods.
Collapse
Affiliation(s)
- Desislava K. Tsoneva
- Department of Medical Genetics, Medical University of Varna, Varna, Bulgaria
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, Varna, Bulgaria
| | - Martin N. Ivanov
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute, Medical University of Varna, Varna, Bulgaria
| | - Manlio Vinciguerra
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, Varna, Bulgaria
- Faculty of Health, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
14
|
Fernandez‐Becerra C, Xander P, Alfandari D, Dong G, Aparici‐Herraiz I, Rosenhek‐Goldian I, Shokouhy M, Gualdron‐Lopez M, Lozano N, Cortes‐Serra N, Karam PA, Meneghetti P, Madeira RP, Porat Z, Soares RP, Costa AO, Rafati S, da Silva A, Santarém N, Fernandez‐Prada C, Ramirez MI, Bernal D, Marcilla A, Pereira‐Chioccola VL, Alves LR, Portillo HD, Regev‐Rudzki N, de Almeida IC, Schenkman S, Olivier M, Torrecilhas AC. Guidelines for the purification and characterization of extracellular vesicles of parasites. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e117. [PMID: 38939734 PMCID: PMC11080789 DOI: 10.1002/jex2.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/21/2023] [Accepted: 09/14/2023] [Indexed: 06/29/2024]
Abstract
Parasites are responsible for the most neglected tropical diseases, affecting over a billion people worldwide (WHO, 2015) and accounting for billions of cases a year and responsible for several millions of deaths. Research on extracellular vesicles (EVs) has increased in recent years and demonstrated that EVs shed by pathogenic parasites interact with host cells playing an important role in the parasite's survival, such as facilitation of infection, immunomodulation, parasite adaptation to the host environment and the transfer of drug resistance factors. Thus, EVs released by parasites mediate parasite-parasite and parasite-host intercellular communication. In addition, they are being explored as biomarkers of asymptomatic infections and disease prognosis after drug treatment. However, most current protocols used for the isolation, size determination, quantification and characterization of molecular cargo of EVs lack greater rigor, standardization, and adequate quality controls to certify the enrichment or purity of the ensuing bioproducts. We are now initiating major guidelines based on the evolution of collective knowledge in recent years. The main points covered in this position paper are methods for the isolation and molecular characterization of EVs obtained from parasite-infected cell cultures, experimental animals, and patients. The guideline also includes a discussion of suggested protocols and functional assays in host cells.
Collapse
Affiliation(s)
- Carmen Fernandez‐Becerra
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
- IGTP Institut d'Investigació Germans Trias i PujolBadalona (Barcelona)Spain
- CIBERINFECISCIII‐CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos IIIMadridSpain
| | - Patrícia Xander
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Daniel Alfandari
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - George Dong
- The Research Institute of the McGill University Health CentreMcGill UniversityMontréalQuébecCanada
| | - Iris Aparici‐Herraiz
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | | | - Mehrdad Shokouhy
- Department of Immunotherapy and Leishmania Vaccine ResearchPasteur Institute of IranTehranIran
| | - Melisa Gualdron‐Lopez
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | - Nicholy Lozano
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Nuria Cortes‐Serra
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | - Paula Abou Karam
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - Paula Meneghetti
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Rafael Pedro Madeira
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Ziv Porat
- Flow Cytometry UnitLife Sciences Core Facilities, WISRehovotIsrael
| | | | - Adriana Oliveira Costa
- Departamento de Análises Clínicas e ToxicológicasFaculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG)Belo HorizonteMinas GeraisBrasil
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine ResearchPasteur Institute of IranTehranIran
| | - Anabela‐Cordeiro da Silva
- Host‐Parasite Interactions GroupInstitute of Research and Innovation in HealthUniversity of PortoPortoPortugal
- Department of Biological SciencesFaculty of PharmacyUniversity of PortoPortoPortugal
| | - Nuno Santarém
- Host‐Parasite Interactions GroupInstitute of Research and Innovation in HealthUniversity of PortoPortoPortugal
- Department of Biological SciencesFaculty of PharmacyUniversity of PortoPortoPortugal
| | | | - Marcel I. Ramirez
- EVAHPI ‐ Extracellular Vesicles and Host‐Parasite Interactions Research Group Laboratório de Biologia Molecular e Sistemática de TripanossomatideosInstituto Carlos Chagas‐FiocruzCuritibaParanáBrasil
| | - Dolores Bernal
- Departament de Bioquímica i Biologia Molecular, Facultat de Ciències BiològiquesUniversitat de ValènciaBurjassotValenciaSpain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i ParasitologiaUniversitat de ValènciaBurjassotValenciaSpain
| | - Vera Lucia Pereira‐Chioccola
- Laboratório de Biologia Molecular de Parasitas e Fungos, Centro de Parasitologia e MicologiaInstituto Adolfo Lutz (IAL)São PauloBrasil
| | - Lysangela Ronalte Alves
- Laboratório de Regulação da Expressão GênicaInstituto Carlos ChagasFiocruz ParanáCuritibaBrazil
- Research Center in Infectious DiseasesDivision of Infectious Disease and Immunity CHU de Quebec Research CenterDepartment of MicrobiologyInfectious Disease and ImmunologyFaculty of MedicineUniversity LavalQuebec CityQuebecCanada
| | - Hernando Del Portillo
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
- IGTP Institut d'Investigació Germans Trias i PujolBadalona (Barcelona)Spain
- ICREA Institució Catalana de Recerca i Estudis Avanc¸ats (ICREA)BarcelonaSpain
| | - Neta Regev‐Rudzki
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - Igor Correia de Almeida
- Department of Biological SciencesBorder Biomedical Research CenterThe University of Texas at El PasoEl PasoTexasUSA
| | - Sergio Schenkman
- Departamento de MicrobiologiaImunologia e Parasitologia, UNIFESPSão PauloBrazil
| | - Martin Olivier
- The Research Institute of the McGill University Health CentreMcGill UniversityMontréalQuébecCanada
| | - Ana Claudia Torrecilhas
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| |
Collapse
|
15
|
Zhang W, Zhang J, Shi H, Liu F, Yu H, Shi H. Exosome GLUT1 derived from hepatocyte identifies the risk of non-alcoholic steatohepatitis and fibrosis. Hepatol Int 2023; 17:1170-1181. [PMID: 37278869 DOI: 10.1007/s12072-023-10520-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/11/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND AND AIMS It is particularly important to identify the progression of non-alcoholic fatty liver disease (NAFLD) for prognosis evaluation and treatment guidance. The aim of this study was to explore the clinic use of exosomal protein-based detection as a valuable non-invasive diagnostic method for NAFLD. METHODS Exosomes were extracted from plasma of patients with NAFLD using Optima XPN-100 ultrafast centrifuge. The patients were recruited from outpatients and inpatients of Beijing Youan Hospital Affiliated to Capital Medical University. The exosomes were stained with fluorescent-labeled antibody and determined by ImageStream® X MKII imaging flow cytometry. Generalized linear logistic regression model was used to evaluate the diagnostic value of hepatogenic exosomes in NAFLD and liver fibrosis. RESULTS The percentage of hepatogenic exosomes glucose transporter 1 (GLUT1) in patients with non-alcoholic steatohepatitis (NASH) was significantly higher than that in patients with non-alcoholic fatty liver (NAFL). According to liver biopsy, we found that the percentage of hepatogenic exosomes GLUT1 in patients with advanced NASH (F2-4) was significantly higher than that in patients with early NASH (F0-1), and the same trend was observed in exosomes with CD63 and ALB. Compared with other clinical fibrosis scoring criteria (FIB-4, NFS, etc.), the diagnostic performance of hepatogenic exosomes GLUT1 was the highest and the area under the receiver-operating curves (AUROC) was 0.85 (95% CI 0.77-0.93). Furthermore, the AUROC of hepatogenic exosomes GLUT1 combined with fibrosis scoring was as high as 0.86-0.91. CONCLUSION Hepatogenic exosome GLUT1 can be a molecular biomarker for early warning of NAFLD to distinguish the NAFL and NASH, and it also can be used as a novel non-invasive diagnostic biomarker for the staging liver fibrosis in NAFLD.
Collapse
Affiliation(s)
- Wenyan Zhang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- State Clinical Drug Trial Institute, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Department of Infectious Diseases, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450000, China
| | - Jing Zhang
- The Third Unit, Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Honglin Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Fang Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Haibin Yu
- State Clinical Drug Trial Institute, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Hongbo Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China.
| |
Collapse
|
16
|
Heidegger S, Stritzke F, Dahl S, Daßler-Plenker J, Joachim L, Buschmann D, Fan K, Sauer CM, Ludwig N, Winter C, Enssle S, Li S, Perl M, Görgens A, Haas T, Orberg ET, Göttert S, Wölfel C, Engleitner T, Cortés-Ciriano I, Rad R, Herr W, Giebel B, Ruland J, Bassermann F, Coch C, Hartmann G, Poeck H. Targeting nucleic acid sensors in tumor cells to reprogram biogenesis and RNA cargo of extracellular vesicles for T cell-mediated cancer immunotherapy. Cell Rep Med 2023; 4:101171. [PMID: 37657445 PMCID: PMC10518594 DOI: 10.1016/j.xcrm.2023.101171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/04/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023]
Abstract
Tumor-derived extracellular vesicles (EVs) have been associated with immune evasion and tumor progression. We show that the RNA-sensing receptor RIG-I within tumor cells governs biogenesis and immunomodulatory function of EVs. Cancer-intrinsic RIG-I activation releases EVs, which mediate dendritic cell maturation and T cell antitumor immunity, synergizing with immune checkpoint blockade. Intact RIG-I, autocrine interferon signaling, and the GTPase Rab27a in tumor cells are required for biogenesis of immunostimulatory EVs. Active intrinsic RIG-I signaling governs composition of the tumor EV RNA cargo including small non-coding stimulatory RNAs. High transcriptional activity of EV pathway genes and RIG-I in melanoma samples associate with prolonged patient survival and beneficial response to immunotherapy. EVs generated from human melanoma after RIG-I stimulation induce potent antigen-specific T cell responses. We thus define a molecular pathway that can be targeted in tumors to favorably alter EV immunomodulatory function. We propose "reprogramming" of tumor EVs as a personalized strategy for T cell-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Simon Heidegger
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.
| | - Florian Stritzke
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sarah Dahl
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Juliane Daßler-Plenker
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Laura Joachim
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Dominik Buschmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| | - Kaiji Fan
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Carolin M Sauer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge, UK
| | - Nils Ludwig
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Christof Winter
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Stefan Enssle
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Suqi Li
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Markus Perl
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - André Görgens
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden; Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tobias Haas
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Erik Thiele Orberg
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sascha Göttert
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Catherine Wölfel
- Internal Medicine III, University Cancer Center and Research Center for Immunotherapy, University Medical Center Johannes Gutenberg University and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge, UK
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany; Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jürgen Ruland
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Coch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology (CITO), Regensburg, Germany.
| |
Collapse
|
17
|
Chae CW, Choi G, Kim YJ, Cho M, Kwon YW, Kim HS. The maintenance mechanism of hematopoietic stem cell dormancy: role for a subset of macrophages. BMB Rep 2023; 56:482-487. [PMID: 37574807 PMCID: PMC10547972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023] Open
Abstract
Hematopoiesis is regulated by crosstalk between long-term repopulating hematopoietic stem cells (LT-HSCs) and supporting niche cells in the bone marrow (BM). Here, we describe the role of KAI1, which is mainly expressed on LT-HSCs and rarely on other hematopoietic stem-progenitor cells (HSPCs), in nichemediated LT-HSC maintenance. KAI1 activates TGF-β1/Smad3 signal in LT-HSCs, leading to the induction of CDK inhibitors and inhibition of the cell cycle. The KAI1-binding partner DARC is expressed on macrophages and stabilizes KAI1 on LT-HSCs, promoting their quiescence. Conversely, when DARC+ BM macrophages were absent, the level of surface KAI1 on LT-HSCs decreases, leading to cell-cycle entry, proliferation, and differentiation. Thus, KAI1 acts as a functional surface marker of LTHSCs that regulates dormancy through interaction with DARCexpressing macrophages in the BM stem cell niche. Recently, we showed very special and rare macrophages expressing α-SMA+ COX2+ & DARC+ induce not only dormancy of LTHSC through interaction of KAI1-DARC but also protect HSCs by down-regulating ROS through COX2 signaling. In the near future, the strategy to combine KAI1-positive LT-HSCs and α-SMA/Cox2/DARC triple-positive macrophages will improve the efficacy of stem cell transplantation after the ablative chemo-therapy for hematological disorders including leukemia. [BMB Reports 2023; 56(9): 482-487].
Collapse
Affiliation(s)
- Cheong-Whan Chae
- Strategic Center of Cell and Bio Therapy for Heart, Diabetes & Cancer, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Gun Choi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
| | - You Ji Kim
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Mingug Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
| | - Yoo-Wook Kwon
- Strategic Center of Cell and Bio Therapy for Heart, Diabetes & Cancer, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyo-Soo Kim
- Strategic Center of Cell and Bio Therapy for Heart, Diabetes & Cancer, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
18
|
Pang K, Dong S, Zhu Y, Zhu X, Zhou Q, Gu B, Jin W, Zhang R, Fu Y, Yu B, Sun D, Duanmu Z, Wei X. Advanced flow cytometry for biomedical applications. JOURNAL OF BIOPHOTONICS 2023; 16:e202300135. [PMID: 37263969 DOI: 10.1002/jbio.202300135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/03/2023]
Abstract
Flow cytometry (FC) is a versatile tool with excellent capabilities to detect and measure multiple characteristics of a population of cells or particles. Notable advancements in in vivo photoacoustic FC, coherent Raman FC, microfluidic FC, and so on, have been achieved in the last two decades, which endows FC with new functions and expands its applications in basic research and clinical practice. Advanced FC broadens the tools available to researchers to conduct research involving cancer detection, microbiology (COVID-19, HIV, bacteria, etc.), and nucleic acid analysis. This review presents an overall picture of advanced flow cytometers and provides not only a clear understanding of their mechanisms but also new insights into their practical applications. We identify the latest trends in this area and aim to raise awareness of advanced techniques of FC. We hope this review expands the applications of FC and accelerates its clinical translation.
Collapse
Affiliation(s)
- Kai Pang
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Sihan Dong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuxi Zhu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xi Zhu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Quanyu Zhou
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Bobo Gu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Jin
- International Cancer Institute, Peking University, Beijing, China
| | - Rui Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuting Fu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Bingchen Yu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Da Sun
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Zheng Duanmu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xunbin Wei
- International Cancer Institute, Peking University, Beijing, China
| |
Collapse
|
19
|
Wu S, Benny M, Duara J, Williams K, Tan A, Schmidt A, Young KC. Extracellular vesicles: pathogenic messengers and potential therapy for neonatal lung diseases. Front Pediatr 2023; 11:1205882. [PMID: 37397144 PMCID: PMC10311919 DOI: 10.3389/fped.2023.1205882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of nano-sized membranous structures increasingly recognized as mediators of intercellular and inter-organ communication. EVs contain a cargo of proteins, lipids and nucleic acids, and their cargo composition is highly dependent on the biological function of the parental cells. Their cargo is protected from the extracellular environment by the phospholipid membrane, thus allowing for safe transport and delivery of their intact cargo to nearby or distant target cells, resulting in modification of the target cell's gene expression, signaling pathways and overall function. The highly selective, sophisticated network through which EVs facilitate cell signaling and modulate cellular processes make studying EVs a major focus of interest in understanding various biological functions and mechanisms of disease. Tracheal aspirate EV-miRNA profiling has been suggested as a potential biomarker for respiratory outcome in preterm infants and there is strong preclinical evidence showing that EVs released from stem cells protect the developing lung from the deleterious effects of hyperoxia and infection. This article will review the role of EVs as pathogenic messengers, biomarkers, and potential therapies for neonatal lung diseases.
Collapse
Affiliation(s)
- Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - Joanne Duara
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - Kevin Williams
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - April Tan
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
| | - Karen C. Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Holtz Children’s Hospital, Jackson Memorial Medical Center, Miami, FL, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
20
|
Lisi V, Senesi G, Bertola N, Pecoraro M, Bolis S, Gualerzi A, Picciolini S, Raimondi A, Fantini C, Moretti E, Parisi A, Sgrò P, Di Luigi L, Geiger R, Ravera S, Vassalli G, Caporossi D, Balbi C. Plasma-derived extracellular vesicles released after endurance exercise exert cardioprotective activity through the activation of antioxidant pathways. Redox Biol 2023; 63:102737. [PMID: 37236143 DOI: 10.1016/j.redox.2023.102737] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases (CVD) can cause various conditions, including an increase in reactive oxygen species (ROS) levels that can decrease nitric oxide (NO) availability and promote vasoconstriction, leading to arterial hypertension. Physical exercise (PE) has been found to be protective against CVD by helping to maintain redox homeostasis through a decrease in ROS levels, achieved by increased expression of antioxidant enzymes (AOEs) and modulation of heat shock proteins (HSPs). Extracellular vesicles (EVs) circulating in the body are a major source of regulatory signals, including proteins and nucleic acids. Interestingly, the cardioprotective role of EVs released after PE has not been fully described. The aim of this study was to investigate the role of circulating EVs, obtained through Size Exclusion Chromatography (SEC) of plasma samples from healthy young males (age: 26.95 ± 3.07; estimated maximum oxygen consumption rate (VO2max): 51.22 ± 4.85 (mL/kg/min)) at basal level (Pre_EVs) and immediately after a single bout of endurance exercise (30' treadmill, 70% heart rate (HR) -Post_EVs). Gene ontology (GO) analysis of proteomic data from isolated EVs, revealed enrichment in proteins endowed with catalytic activity in Post_EVs, compare to Pre_EVs, with MAP2K1 being the most significantly upregulated protein. Enzymatic assays on EVs derived from Pre and Post samples showed increment in Glutathione Reductase (GR) and Catalase (CAT) activity in Post_EVs. At functional level, Post_EVs, but not Pre_EVs, enhanced the activity of antioxidant enzymes (AOEs) and reduced oxidative damage accumulation in treated human iPS-derived cardiomyocytes (hCM) at basal level and under stress conditions (Hydrogen Peroxide (H2O2) treatment), resulting in a global cardioprotective effect. In conclusion, our data demonstrated, for the first time, that a single 30-min endurance exercise is able to alter the cargo of circulating EVs, resulting in cardioprotective effect through antioxidant activity.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, 16132, Genova, Italy
| | - Matteo Pecoraro
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Alice Gualerzi
- Laboratory of Nanomedicine and Clinical Biophotonics (LABION), IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Silvia Picciolini
- Laboratory of Nanomedicine and Clinical Biophotonics (LABION), IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Andrea Raimondi
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland; Centro Imaging Sperimentale, IRCCS Istituto Scientifico San Raffaele, Via Olgettina 52, 20132, Milan, Italy
| | - Cristina Fantini
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Elisa Moretti
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Attilio Parisi
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Paolo Sgrò
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Luigi Di Luigi
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132, Genova, Italy
| | - Giuseppe Vassalli
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland.
| |
Collapse
|
21
|
Thompson W, Papoutsakis ET. The role of biomechanical stress in extracellular vesicle formation, composition and activity. Biotechnol Adv 2023; 66:108158. [PMID: 37105240 DOI: 10.1016/j.biotechadv.2023.108158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Extracellular vesicles (EVs) are cornerstones of intercellular communication with exciting fundamental, clinical, and more broadly biotechnological applications. However, variability in EV composition, which results from the culture conditions used to generate the EVs, poses significant fundamental and applied challenges and a hurdle for scalable bioprocessing. Thus, an understanding of the relationship between EV production (and for clinical applications, manufacturing) and EV composition is increasingly recognized as important and necessary. While chemical stimulation and culture conditions such as cell density are known to influence EV biology, the impact of biomechanical forces on the generation, properties, and biological activity of EVs remains poorly understood. Given the omnipresence of these forces in EV preparation and in biomanufacturing, expanding the understanding of their impact on EV composition-and thus, activity-is vital. Although several publications have examined EV preparation and bioprocessing and briefly discussed biomechanical stresses as variables of interest, this review represents the first comprehensive evaluation of the impact of such stresses on EV production, composition and biological activity. We review how EV biogenesis, cargo, efficacy, and uptake are uniquely affected by various types, magnitudes, and durations of biomechanical forces, identifying trends that emerge both generically and for individual cell types. We also describe implications for scalable bioprocessing, evaluating processes inherent in common EV production and isolation methods, and propose a path forward for rigorous EV quality control.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA.
| |
Collapse
|
22
|
Tsoneva DK, Ivanov MN, Conev NV, Manev R, Stoyanov DS, Vinciguerra M. Circulating Histones to Detect and Monitor the Progression of Cancer. Int J Mol Sci 2023; 24:ijms24020942. [PMID: 36674455 PMCID: PMC9860657 DOI: 10.3390/ijms24020942] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Liquid biopsies have emerged as a minimally invasive cancer detection and monitoring method, which could identify cancer-related alterations in nucleosome or histone levels and modifications in blood, saliva, and urine. Histones, the core component of the nucleosome, are essential for chromatin compaction and gene expression modulation. Increasing evidence suggests that circulating histones and histone complexes, originating from cell death or immune cell activation, could act as promising biomarkers for cancer detection and management. In this review, we provide an overview of circulating histones as a powerful liquid biopsy approach and methods for their detection. We highlight current knowledge on circulating histones in hematologic malignancies and solid cancer, with a focus on their role in cancer dissemination, monitoring, and tumorigenesis. Last, we describe recently developed strategies to identify cancer tissue-of-origin in blood plasma based on nucleosome positioning, inferred from nucleosomal DNA fragmentation footprint, which is independent of the genetic landscape.
Collapse
Affiliation(s)
- Desislava K. Tsoneva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Martin N. Ivanov
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Nikolay Vladimirov Conev
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Rostislav Manev
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Dragomir Svetozarov Stoyanov
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Manlio Vinciguerra
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
- Correspondence:
| |
Collapse
|
23
|
De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1835. [PMID: 35898167 PMCID: PMC10078256 DOI: 10.1002/wnan.1835] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles (EVs) are a unique and heterogeneous class of lipid bilayer nanoparticles secreted by most cells. EVs are regarded as important mediators of intercellular communication in both prokaryotic and eukaryotic cells due to their ability to transfer proteins, lipids and nucleic acids to recipient cells. In addition to their physiological role, EVs are recognized as modulators in pathological processes such as cancer, infectious diseases, and neurodegenerative disorders, providing new potential targets for diagnosis and therapeutic intervention. For a complete understanding of EVs as a universal cellular biological system and its translational applications, optimal techniques for their isolation and characterization are required. Here, we review recent progress in those techniques, from isolation methods to characterization techniques. With interest in therapeutic applications of EVs growing, we address fundamental points of EV-related cell biology, such as cellular uptake mechanisms and their biodistribution in tissues as well as challenges to their application as drug carriers or biomarkers for less invasive diagnosis or as immunogens. This article is categorized under: Diagnostic Tools > Biosensing Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Karina P. De Sousa
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
| | - Izadora Rossi
- School of Human SciencesLondon Metropolitan UniversityLondonUK
- Federal University of ParanáCuritibaBrazil
| | | | - Marcel Ivan Ramirez
- Federal University of ParanáCuritibaBrazil
- Carlos Chagas Institute (ICC)CuritibaBrazil
| | - Dan Stratton
- Open UniversityThe School of Life, Health and Chemical SciencesMilton KeynesUK
| | - Jameel Malhador Inal
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
- School of Human SciencesLondon Metropolitan UniversityLondonUK
| |
Collapse
|
24
|
Panja S, Siegel D, Camandola S, de Cabo R, Ross D, Mallela K. FAD-deficient P187S mutation of NAD(P)H:quinone oxidoreductase 1 (NQO1*2) binds and accelerates β-amyloid aggregation. Biosci Rep 2022; 42:BSR20220643. [PMID: 36281795 PMCID: PMC9664297 DOI: 10.1042/bsr20220643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/28/2022] [Accepted: 10/24/2022] [Indexed: 02/03/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most prominent neurodegenerative diseases. Results from animal and cellular models suggest that FAD-deficient forms of NAD(P)H quinone oxidoreductase 1 (NQO1) may accelerate the aggregation of Alzheimer's amyloid-β peptide (Aβ1-42). Here, we examined in vitro whether NQO1 and its FAD-deficient P187S mutation (NQO1*2) directly interact with Aβ1-42 and modify its rate of aggregation. When monitored using the fluorescence of either noncovalent thioflavin T (ThT) or HiLyte Fluor 647 (HF647) dye covalently attached to the Aβ1-42 peptide, the aggregation kinetics of Aβ1-42 were markedly more rapid in the presence of NQO1*2 than the wild-type (WT) NQO1. Experiments using apo-NQO1 indicate that this increase is linked to the inability of NQO1*2 to bind to FAD. Furthermore, dicoumarol, an NQO1 inhibitor that binds near the FAD-binding site and stabilizes NQO1*2, markedly decreased the aggregation kinetics of Aβ1-42. Imaging flow cytometry confirmed in-vitro coaggregation of NQO1 isoforms and Aβ1-42. Aβ1-42 alone forms rod-shaped fibril structures while in the presence of NQO1 isoforms, Aβ1-42 is incorporated in the middle of larger globular protein aggregates surrounded by NQO1 molecules. Isothermal titration calorimetry (ITC) analysis indicates that Aβ1-42 interacts with NQO1 isoforms with a specific stoichiometry through a hydrophobic interaction with positive enthalpy and entropy changes. These data define the kinetics, mechanism, and shape of coaggregates of Aβ1-42 and NQO1 isoforms and the potential relevance of FAD-deficient forms of NQO1 for amyloid aggregation diseases.
Collapse
Affiliation(s)
- Sudipta Panja
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, MS C238-V20, Aurora, CO 80045, U.S.A
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, MS C238-V20, Aurora, CO 80045, U.S.A
| | - Simonetta Camandola
- Experimental Gerontology Section, National Institute of Aging, National Institutes of Health, Baltimore MD, U.S.A
| | - Rafael de Cabo
- Experimental Gerontology Section, National Institute of Aging, National Institutes of Health, Baltimore MD, U.S.A
| | - David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, MS C238-V20, Aurora, CO 80045, U.S.A
| | - Krishna M.G. Mallela
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, MS C238-V20, Aurora, CO 80045, U.S.A
| |
Collapse
|
25
|
Marki A, Ley K. The expanding family of neutrophil-derived extracellular vesicles. Immunol Rev 2022; 312:52-60. [PMID: 35665941 PMCID: PMC10111154 DOI: 10.1111/imr.13103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022]
Abstract
Neutrophils are immune cells involved in several inflammatory and homeostatic processes. Their capacity to release cargo can be classified based on whether the cargo is released on its own, or in conjunction with plasma membrane structures. Examples of plasma membrane-free secretion modes are degranulation, neutrophil extracellular trap (NET) release, and cytokine release through inflammasome formation. The most studied membrane-covered neutrophil-derived structures are exosomes and ectosomes that are collectively called extracellular vesicles (EV). Apoptotic vesicles are another recognized EV subtype. Over the last decade, additional membrane-covered neutrophil-derived structures were characterized: migratory cytoplasts, migrasomes, and elongated neutrophil-derived structures (ENDS). All these structures are smaller than the neutrophils, cannot reproduce themselves, and thus meet the latest consensus definition of EVs. In this review, we focus on the less well-studied neutrophil EVs: apoptotic vesicles, cytoplasts, migrasomes, and ENDS.
Collapse
Affiliation(s)
- Alex Marki
- AstraZeneca, Gaithersburg, Maryland, USA
| | - Klaus Ley
- La Jolla Institute for Immunology and Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
26
|
Dandugudumula R, Fischer-Weinberger R, Zilberstein D. Morphogenesis Dynamics in Leishmania Differentiation. Pathogens 2022; 11:952. [PMID: 36145385 PMCID: PMC9505065 DOI: 10.3390/pathogens11090952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Leishmania, the causative agent of leishmaniasis, is an obligatory intracellular parasite that cycles between phagolysosome of mammalian macrophages, where it resides as round intracellular amastigotes, and the midgut of female sandflies, where it resides as extracellular elongated promastigotes. This protozoan parasite cytoskeleton is composed of stable and abundant subpellicular microtubules (SPMT). This study aims to determine the kinetics of developmental morphogenesis and assess whether microtubules remodelling is involved in this process. Using image-streaming technology, we observed that rounding of promastigotes during differentiation into amastigotes was initiated promptly after exposure to the differentiation signal. Stabilizing microtubules with taxol sped rounding, but later killed differentiating parasites if taxol was not removed. Microtubule destabilizers such as vinblastine had no effect on the rate of rounding, nor on the viability of differentiating parasites. In the reverse process, elongation is initiated after a delay of 7.5 and completed 72 h after exposure to the amastigote to the promastigote differentiation signal. During the delay, parasites became highly sensitive to treatment with microtubule destabilizers. The addition of vinblastine during the first 7.5 h halted differentiation and killed parasites. Between 8 and 24 h, parasites gradually became resistant to vinblastine and, in parallel, started to elongate. In contrast, taxol had no effect on parasite elongation, nor on the viability of these cells. In a parallel study, we showed that the Leishmania-specific protein kinase A (PKA) holoenzyme containing the LdPKAR3-C3 complex is essential for promastigote elongation. Mutant promastigotes lacking either of these proteins are round, but maintain their flagella. Here, we observed that during differentiation into amastigotes, these mutants round at the same rate as the wild type, but never exceed the WT density of round amastigotes. In the reverse process, these mutants undergo the same initial delay and then elongate at the same rate as the WT. They stop elongating when they reach 20% of elongated cells in mature promastigotes. Our analysis indicates that while promastigote rounding into amastigotes did not require microtubule remodelling, morphogenesis of round amastigotes into elongated promastigotes required microtubule rearrangement before elongation was initiated. This is the first study that investigates the dynamics of microtubules during parasite development.
Collapse
Affiliation(s)
| | | | - Dan Zilberstein
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
27
|
Wu J, Wu D, Wu G, Bei HP, Li Z, Xu H, Wang Y, Wu D, Liu H, Shi S, Zhao C, Xu Y, He Y, Li J, Wang C, Zhao X, Wang S. Scale-out production of extracellular vesicles derived from natural killer cells via mechanical stimulation in a seesaw-motion bioreactor for cancer therapy. Biofabrication 2022; 14. [PMID: 35793612 DOI: 10.1088/1758-5090/ac7eeb] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/06/2022] [Indexed: 11/11/2022]
Abstract
Extracellular vesicles (EVs) derived from immune cells have shown great anti-cancer therapeutic potential. However, inefficiency in EV generation has considerably impeded the development of EV-based basic research and clinical translation. Here, we developed a seesaw-motion bioreactor (SMB) system by leveraging mechanical stimuli such as shear stress and turbulence for generating EVs with high quality and quantity from natural killer (NK) cells. Compared to EV production in traditional static culture (229 ± 74 particles per cell per day), SMB produced NK-92MI-derived EVs at a higher rate of 438 ± 50 particles per cell per day and yielded a total number of 2 × 1011 EVs over two weeks via continuous dynamic fluidic culture. In addition, the EVs generated from NK-92MI cells in SMB shared a similar morphology, size distribution, and protein profile to EVs generated from traditional static culture. Most importantly, the NK-92MI-derived EVs in SMB were functionally active in killing melanoma and liver cancer cells in both 2D and 3D culture conditions in vitro, as well as in suppressing melanoma growth in vivo. We believe that SMB is an attractive approach to producing EVs with high quality and quantity; it can additionally enhance EV production from NK92-MI cells and promote both the basic and translational research of EVs.
Collapse
Affiliation(s)
- Jianguo Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Di Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Guohua Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong, Hong Kong SAR, HONG KONG
| | - Zihan Li
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Han Xu
- Department of Building Environment and Energy Engineering, Xi'an Jiaotong University, 28 Xianning W Rd, Beilin, Xi'An, Shaanxi, China, 710049, Xi'an, Shanxi Province, 710049, CHINA
| | - Yimin Wang
- Institute of Translational Medicine, Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, HangZhou, 310027, CHINA
| | - Dan Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Hui Liu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Shengyu Shi
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, The Old Schools, Trinity Ln, Cambridge CB2 1TN, United Kingdom, Cambridge, CB2 1TN, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Yibing Xu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Yong He
- Department of Mechanical Engineering, Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, ZheJiang, 310027, CHINA
| | - Jun Li
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Changyong Wang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institude of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Academy of Military Medical Sciences, Taiping Rd. 27, 100850, Tianjin, Beijing, China, Beijing, 100850, CHINA
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong, Hong Kong SAR, 999077, HONG KONG
| | - Shuqi Wang
- Sichuan University, 252 Shuncheng Ave, Qingyang District, Chengdu, Sichuan, China, Chengdu, Sichuan, 610017, CHINA
| |
Collapse
|
28
|
Woud WW, van der Pol E, Mul E, Hoogduijn MJ, Baan CC, Boer K, Merino A. An imaging flow cytometry-based methodology for the analysis of single extracellular vesicles in unprocessed human plasma. Commun Biol 2022; 5:633. [PMID: 35768629 PMCID: PMC9243126 DOI: 10.1038/s42003-022-03569-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/18/2022] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles (EVs) are tissue-specific particles released by cells containing valuable diagnostic information in the form of various biomolecules. To rule out selection bias or introduction of artefacts caused by EV isolation techniques, we present a clinically feasible, imaging flow cytometry (IFCM)-based methodology to phenotype and determine the concentration of EVs with a diameter ≤400 nm in human platelet-poor plasma (PPP) without prior isolation of EVs. Instrument calibration (both size and fluorescence) were performed with commercial polystyrene beads. Detergent treatment of EVs was performed to discriminate true vesicular events from artefacts. Using a combination of markers (CFSE & Tetraspanins, or CD9 & CD31) we found that >90% of double-positive fluorescent events represented single EVs. Through this work, we provide a framework that will allow the application of IFCM for EV analysis in peripheral blood plasma in a plethora of experimental and potentially diagnostic settings. Additionally, this direct approach for EV analysis will enable researchers to explore corners of EVs as cellular messengers in healthy and pathological conditions.
Collapse
Affiliation(s)
- Wouter W Woud
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Edwin van der Pol
- Biomedical Engineering & Physics, Laboratory Experimental Clinical Chemistry, Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Erik Mul
- Department Central Cell Analysis Facility, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin J Hoogduijn
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carla C Baan
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karin Boer
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ana Merino
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Abstract
Extracellular vesicles (EVs) are membranous nanoparticles secreted by nearly all cell types and play a critical role in cell-to-cell crosstalk. EVs can be categorized based on their size, surface markers, or the cell type from which they originate. EVs carry "cargo," including but not limited to, RNA, DNA, proteins, and small signaling molecules. To date, many methods have been developed to isolate EVs from biological fluids, such as blood plasma, urine, bronchoalveolar lavage fluid, and urine. Once isolated, EVs can be characterized by dynamic light scattering, nanotracking analysis, nanoscale flow cytometry, and transmission electron microscopy. Given the ability of EVs to transport cargo between cells, research has recently focused on understanding their role in various human diseases. As understanding of their significance to disease processes grows, insight into the mechanisms behind the physiological role of their cargo in target cells can facilitate the development of a new type of biomarker and therapeutic target for diseases in future. In addition, their ability to deliver their cargo selectively to target cells within the human body means that they could serve as therapeutic agents or methods of drug delivery. In this review, we will first introduce EVs and the cargo they carry, outline current methods for EV isolation and characterization, and discuss their potential use as biomarkers and therapeutic agents in the near future.
Collapse
Affiliation(s)
- Jonathan M Carnino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA, United States
| | - Heedoo Lee
- Department of Biology and Chemistry, Changwon National University, Changwon, South Korea.
| |
Collapse
|
30
|
Dunker S, Boyd M, Durka W, Erler S, Harpole WS, Henning S, Herzschuh U, Hornick T, Knight T, Lips S, Mäder P, Švara EM, Mozarowski S, Rakosy D, Römermann C, Schmitt‐Jansen M, Stoof‐Leichsenring K, Stratmann F, Treudler R, Virtanen R, Wendt‐Potthoff K, Wilhelm C. The potential of multispectral imaging flow cytometry for environmental monitoring. Cytometry A 2022; 101:782-799. [DOI: 10.1002/cyto.a.24658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/23/2022] [Accepted: 05/12/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Susanne Dunker
- Department of Physiological Diversity Helmholtz‐Centre for Environmental Research (UFZ) Leipzig Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
| | - Matthew Boyd
- Department of Anthropology Lakehead University Thunder Bay Canada
| | - Walter Durka
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Department of Community Ecology Helmholtz‐Centre for Environmental Research (UFZ) Halle Germany
| | - Silvio Erler
- Institute for Bee Protection, Julius Kühn Institute (JKI)‐Federal Research Centre for Cultivated Plants Braunschweig Germany
| | - W. Stanley Harpole
- Department of Physiological Diversity Helmholtz‐Centre for Environmental Research (UFZ) Leipzig Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Institute of Biology, Martin Luther University Halle‐Wittenberg Halle Germany
| | - Silvia Henning
- Department of Experimental Aerosol and Cloud Microphysics Leibniz Institute for Tropospheric Research (TROPOS) Leipzig Germany
| | - Ulrike Herzschuh
- Alfred‐Wegner‐Institute Helmholtz Centre of Polar and Marine Research Polar Terrestrial Environmental Systems Potsdam Germany
- Institute of Environmental Sciences and Geography University of Potsdam Potsdam Germany
- Institute of Biochemistry and Biology University of Potsdam Potsdam Germany
| | - Thomas Hornick
- Department of Physiological Diversity Helmholtz‐Centre for Environmental Research (UFZ) Leipzig Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
| | - Tiffany Knight
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Department of Community Ecology Helmholtz‐Centre for Environmental Research (UFZ) Halle Germany
- Institute of Biology, Martin Luther University Halle‐Wittenberg Halle Germany
| | - Stefan Lips
- Department of Bioanalytical Ecotoxicology Helmholtz‐Centre for Environmental Research – UFZ Leipzig Germany
| | - Patrick Mäder
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Department of Computer Science and Automation Technische Universität Ilmenau Ilmenau Germany
- Faculty of Biological Sciences Friedrich‐Schiller‐University Jena Jena Germany
| | - Elena Motivans Švara
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Department of Community Ecology Helmholtz‐Centre for Environmental Research (UFZ) Halle Germany
- Institute of Biology, Martin Luther University Halle‐Wittenberg Halle Germany
| | | | - Demetra Rakosy
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Department of Community Ecology Helmholtz‐Centre for Environmental Research (UFZ) Halle Germany
| | - Christine Römermann
- German Centre for Integrative Biodiversity Research (iDiv) Halle‐Jena‐Leipzig Leipzig Germany
- Institute of Ecology and Evolution Friedrich‐Schiller‐University Jena Jena Germany
| | - Mechthild Schmitt‐Jansen
- Department of Bioanalytical Ecotoxicology Helmholtz‐Centre for Environmental Research – UFZ Leipzig Germany
| | - Kathleen Stoof‐Leichsenring
- Alfred‐Wegner‐Institute Helmholtz Centre of Polar and Marine Research Polar Terrestrial Environmental Systems Potsdam Germany
| | - Frank Stratmann
- Department of Experimental Aerosol and Cloud Microphysics Leibniz Institute for Tropospheric Research (TROPOS) Leipzig Germany
| | - Regina Treudler
- Department of Dermatology, Venerology and Allergology University of Leipzig Medical Center Leipzig Germany
| | | | - Katrin Wendt‐Potthoff
- Department of Lake Research Helmholtz‐Centre for Environmental Research – UFZ Magdeburg Germany
| | - Christian Wilhelm
- Faculty of Life Sciences, Institute of Biology University of Leipzig Leipzig Germany
| |
Collapse
|
31
|
Vdovenko D, Balbi C, Di Silvestre D, Passignani G, Puspitasari YM, Zarak-Crnkovic M, Mauri P, Camici GG, Lüscher TF, Eriksson U, Vassalli G. Microvesicles released from activated CD4 + T cells alter microvascular endothelial cell function. Eur J Clin Invest 2022; 52:e13769. [PMID: 35316536 PMCID: PMC9287044 DOI: 10.1111/eci.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/18/2021] [Accepted: 01/02/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Microvesicles are vesicles shed by plasma membranes following cell activation and apoptosis. The role of lymphocyte-derived microvesicles in endothelial function remains poorly understood. METHODS CD4+ T cells isolated from peripheral blood of healthy human donors were stimulated using anti-CD3/anti-CD28-coated beads. Proteomic profiling of microvesicles was performed using linear discriminant analysis (LDA) from activated T cells (MV.Act) and nonactivated T cells (MV.NAct). In addition, data processing analysis was performed using MaxQUANT workflow. Differentially expressed proteins found in MV.Act or MV.NAct samples with identification frequency = 100%, which were selected by both LDA (p < .01) and MaxQUANT (p < .01) workflows, were defined as "high-confidence" differentially expressed proteins. Functional effects of MV.Act on human primary microvascular endothelial cells were analysed. RESULTS T cells released large amounts of microvesicles upon stimulation. Proteomic profiling of microvesicles using LDA identified 2279 proteins (n = 2110 and n = 851 proteins in MV.Act and MV.NAct, respectively). Protein-protein interaction network models reconstructed from both differentially expressed proteins (n = 594; LDA p ≤ .01) and "high-confidence" differentially expressed proteins (n = 98; p ≤ .01) revealed that MV.Act were enriched with proteins related to immune responses, protein translation, cytoskeleton organisation and TNFα-induced apoptosis. For instance, MV.Act were highly enriched with IFN-γ, a key proinflammatory pathway related to effector CD4+ T cells. Endothelial cell incubation with MV.Act induced superoxide generation, apoptosis, endothelial wound healing impairment and endothelial monolayer barrier disruption. CONCLUSIONS T cell receptor-mediated activation of CD4+ T cells stimulates the release of microvesicles enriched with proteins involved in immune responses, inflammation and apoptosis. T cell-derived microvesicles alter microvascular endothelial function and barrier permeability, potentially promoting tissue inflammation.
Collapse
Affiliation(s)
- Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland.,Laboratories for Translational Research-EOC, Bellinzona, Switzerland
| | | | | | | | | | | | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Royal Brompton & Harefield Hospital, Imperial College, London, UK
| | - Urs Eriksson
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Medicine, GZO - Zurich Regional Health Center, Wetzikon, Switzerland
| | - Giuseppe Vassalli
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland.,Laboratories for Translational Research-EOC, Bellinzona, Switzerland.,Department of Biomedicine, Università della Svizzera Italiana (USI), Lugano, Switzerland
| |
Collapse
|
32
|
Stampouloglou PK, Siasos G, Bletsa E, Oikonomou E, Vogiatzi G, Kalogeras K, Katsianos E, Vavuranakis MA, Souvaliotis N, Vavuranakis M. The Role of Cell Derived Microparticles in Cardiovascular Diseases: Current Concepts. Curr Pharm Des 2022; 28:1745-1757. [DOI: 10.2174/1381612828666220429081555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 12/07/2022]
Abstract
Abstract:
Cardiovascular disease remains the main cause of human morbidity and mortality in the developed countries. Microparticles (MPs) are small vesicles originating from the cell membrane as a result of various stimuli and particularly of biological processes that constitute the pathophysiology of atherosclerosis, such as endothelial damage. They form vesicles that can transfer various molecules and signals to remote target cells without direct cell to cell interaction. Circulating microparticles have been associated with cardiovascular diseases. Therefore, many studies have been designed to further investigate the role of microparticles as biomarkers for diagnosis, prognosis, and disease monitoring. To this concept the pro-thrombotic and atherogenic potential of platelets and endothelial derived MPs has gain research interest especially concerning accelerate atherosclerosis and acute coronary syndrome triggering and prognosis. MPs especially of endothelial origin have been investigated in different clinical scenarios of heart failure and in association of left ventricular loading conditions. Finally, most cardiovascular risk factors present unique patterns of circulating MPs population, highlighting their pathophysiologic link to cardiovascular disease progression. In this review article we present a synopsis of the biogenesis and characteristics of microparticles, as well as the most recent data concerning their implication in the cardiovascular settings.
Collapse
Affiliation(s)
- Panagiota K. Stampouloglou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Evanthia Bletsa
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Georgia Vogiatzi
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Efstratios Katsianos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Nektarios Souvaliotis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| |
Collapse
|
33
|
Christopher JA, Geladaki A, Dawson CS, Vennard OL, Lilley KS. Subcellular Transcriptomics and Proteomics: A Comparative Methods Review. Mol Cell Proteomics 2022; 21:100186. [PMID: 34922010 PMCID: PMC8864473 DOI: 10.1016/j.mcpro.2021.100186] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
The internal environment of cells is molecularly crowded, which requires spatial organization via subcellular compartmentalization. These compartments harbor specific conditions for molecules to perform their biological functions, such as coordination of the cell cycle, cell survival, and growth. This compartmentalization is also not static, with molecules trafficking between these subcellular neighborhoods to carry out their functions. For example, some biomolecules are multifunctional, requiring an environment with differing conditions or interacting partners, and others traffic to export such molecules. Aberrant localization of proteins or RNA species has been linked to many pathological conditions, such as neurological, cancer, and pulmonary diseases. Differential expression studies in transcriptomics and proteomics are relatively common, but the majority have overlooked the importance of subcellular information. In addition, subcellular transcriptomics and proteomics data do not always colocate because of the biochemical processes that occur during and after translation, highlighting the complementary nature of these fields. In this review, we discuss and directly compare the current methods in spatial proteomics and transcriptomics, which include sequencing- and imaging-based strategies, to give the reader an overview of the current tools available. We also discuss current limitations of these strategies as well as future developments in the field of spatial -omics.
Collapse
Affiliation(s)
- Josie A Christopher
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Aikaterini Geladaki
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Department of Genetics, University of Cambridge, Cambridge, UK
| | - Charlotte S Dawson
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Owen L Vennard
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
| |
Collapse
|
34
|
Zwozdesky MA, Fei C, Stafford JL. An Imaging Flow Cytometry Protocol for Studying Immunoregulatory Receptor-Mediated Regulation of Phagocytosis. Methods Mol Biol 2022; 2421:201-216. [PMID: 34870821 DOI: 10.1007/978-1-0716-1944-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Advances in flow cytometry have allowed for innovative functional investigations of innate immune cell responses. Imaging flow cytometers combine the imaging capabilities of microscopy with rapid, high-throughput data acquisition attributes of standard flow cytometers. Here, we describe a detailed method for co-expressing stimulatory and inhibitory immunoregulatory receptor-types in AD293 cells and then measuring receptor cross-talk during the regulation of the phagocytic response. Information on reagent selection, imaging flow cytometry calibration, and automated template analyses are included.
Collapse
Affiliation(s)
- Myron A Zwozdesky
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Chenjie Fei
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
35
|
Mochalova EN, Kotov IA, Lifanov DA, Chakraborti S, Nikitin MP. Imaging flow cytometry data analysis using convolutional neural network for quantitative investigation of phagocytosis. Biotechnol Bioeng 2021; 119:626-635. [PMID: 34750809 DOI: 10.1002/bit.27986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/07/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
Macrophages play an important role in the adaptive immune system. Their ability to neutralize cellular targets through Fc receptor-mediated phagocytosis has relied upon immunotherapy that has become of particular interest for the treatment of cancer and autoimmune diseases. A detailed investigation of phagocytosis is the key to the improvement of the therapeutic efficiency of existing medications and the creation of new ones. A promising method for studying the process is imaging flow cytometry (IFC) that acquires thousands of cell images per second in up to 12 optical channels and allows multiparametric fluorescent and morphological analysis of samples in the flow. However, conventional IFC data analysis approaches are based on a highly subjective manual choice of masks and other processing parameters that can lead to the loss of valuable information embedded in the original image. Here, we show the application of a Faster region-based convolutional neural network (CNN) for accurate quantitative analysis of phagocytosis using imaging flow cytometry data. Phagocytosis of erythrocytes by peritoneal macrophages was chosen as a model system. CNN performed automatic high-throughput processing of datasets and demonstrated impressive results in the identification and classification of macrophages and erythrocytes, despite the variety of shapes, sizes, intensities, and textures of cells in images. The developed procedure allows determining the number of phagocytosed cells, disregarding cases with a low probability of correct classification. We believe that CNN-based approaches will enable powerful in-depth investigation of a wide range of biological processes and will reveal the intricate nature of heterogeneous objects in images, leading to completely new capabilities in diagnostics and therapy.
Collapse
Affiliation(s)
- Elizaveta N Mochalova
- Nanobiotechnology Laboratory, Moscow Institute of Physics and Technology, Moscow, Russia.,Biophotonics Laboratory, Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia.,Nanobiomedicine Division, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan A Kotov
- Nanobiotechnology Laboratory, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Dmitry A Lifanov
- Nanobiotechnology Laboratory, Moscow Institute of Physics and Technology, Moscow, Russia
| | | | - Maxim P Nikitin
- Nanobiotechnology Laboratory, Moscow Institute of Physics and Technology, Moscow, Russia.,Nanobiomedicine Division, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
36
|
Acrylate protects a marine bacterium from grazing by a ciliate predator. Nat Microbiol 2021; 6:1351-1356. [PMID: 34697458 DOI: 10.1038/s41564-021-00981-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 09/16/2021] [Indexed: 12/26/2022]
Abstract
Cleavage of dimethylsulfoniopropionate (DMSP) can deter herbivores in DMSP-producing eukaryotic algae; however, it is unclear whether a parallel defence mechanism operates in marine bacteria. Here we demonstrate that the marine bacterium Puniceibacterium antarcticum SM1211, which does not use DMSP as a carbon source, has a membrane-associated DMSP lyase, DddL. At high concentrations of DMSP, DddL causes an accumulation of acrylate around cells through the degradation of DMSP, which protects against predation by the marine ciliate Uronema marinum. The presence of acrylate can alter the grazing preference of U. marinum to other bacteria in the community, thereby influencing community structure.
Collapse
|
37
|
Monnamorn L, Seree-Aphinan C, Molika P, Vichitkunakorn P, Pattanapanyasat K, Khwannimit B, Navakanitworakul R. The Concentration of Large Extracellular Vesicles Differentiates Early Septic Shock From Infection. Front Med (Lausanne) 2021; 8:724371. [PMID: 34604260 PMCID: PMC8481381 DOI: 10.3389/fmed.2021.724371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/24/2021] [Indexed: 11/13/2022] Open
Abstract
Septic shock represents a subset of sepsis with severe physiological aberrations and a higher mortality rate than sepsis alone. Currently, the laboratory tools which can be used to identify the state of septic shock are limited. In pre-clinical studies, extracellular vesicles (EVs), especially large EVs (lEVs), have been demonstrated a role as functional inflammatory mediators of sepsis. However, its longitudinal trend during the disease course has not been explored. In this study, the quantities and subtypes of plasma-derived lEVs were longitudinally compared between patients with septic shock (n = 21) and non-sepsis infection (n = 9), who presented within 48 h of their symptom onset. Blood specimens were collected for seven consecutive days after hospital admission. lEVs quantification and subtyping were performed using an imaging flow cytometer. The experiments revealed a higher lEVs concentration in septic shock patients than infected patients at the onset of the disease. In septic shock patients, lEVs concentration decreased over time as opposed to infected patients whose lEVs concentration is relatively static throughout the study period. The major contributors of lEVs in both septic shock and infected patients were of non-leukocyte origins; platelets, erythrocytes, and endothelial cells released approximately 40, 25, and 15% of lEVs, respectively. Among lEVs of leukocyte origins, neutrophils produced the highest number of EVs. Nevertheless, the proportion of each subtype of lEVs among the given amount of lEVs produced was similar between septic shock and infected patients. These findings raise the possibility of employing lEVs enumeration as a septic shock identifying tool, although larger studies with a more diverse group of participants are warranted to extrapolate the findings to a general population.
Collapse
Affiliation(s)
- Latthawan Monnamorn
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| | - Chutima Seree-Aphinan
- Faculty of Medicine, Department of Internal Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Piyatida Molika
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| | - Polathep Vichitkunakorn
- Faculty of Medicine, Department of Family and Preventive Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Kovit Pattanapanyasat
- Faculty of Medicine Siriraj Hospital, Department of Research and Development, Mahidol University, Bangkok, Thailand
| | - Bodin Khwannimit
- Faculty of Medicine, Department of Internal Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Raphatphorn Navakanitworakul
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
38
|
Zhao M, Zhang F, Zarnowski R, Barns K, Jones R, Fossen J, Sanchez H, Rajski SR, Audhya A, Bugni TS, Andes DR. Turbinmicin inhibits Candida biofilm growth by disrupting fungal vesicle-mediated trafficking. J Clin Invest 2021; 131:145123. [PMID: 33373326 DOI: 10.1172/jci145123] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of drug-resistant fungi has prompted an urgent threat alert from the US Centers for Disease Control (CDC). Biofilm assembly by these pathogens further impairs effective therapy. We recently identified an antifungal, turbinmicin, that inhibits the fungal vesicle-mediated trafficking pathway and demonstrates broad-spectrum activity against planktonically growing fungi. During biofilm growth, vesicles with unique features play a critical role in the delivery of biofilm extracellular matrix components. As these components are largely responsible for the drug resistance associated with biofilm growth, we explored the utility of turbinmicin in the biofilm setting. We found that turbinmicin disrupted extracellular vesicle (EV) delivery during biofilm growth and that this impaired the subsequent assembly of the biofilm matrix. We demonstrated that elimination of the extracellular matrix rendered the drug-resistant biofilm communities susceptible to fungal killing by turbinmicin. Furthermore, the addition of turbinmicin to otherwise ineffective antifungal therapy potentiated the activity of these drugs. The underlying role of vesicles explains this dramatic activity and was supported by phenotype reversal with the addition of exogenous biofilm EVs. This striking capacity to cripple biofilm assembly mechanisms reveals a new approach to eradicating biofilms and sheds light on turbinmicin as a promising anti-biofilm drug.
Collapse
Affiliation(s)
- Miao Zhao
- Department of Medicine, Medical Microbiology and Immunology
| | - Fan Zhang
- Pharmaceutical Sciences Division, and
| | | | | | - Ryley Jones
- Department of Medicine, Medical Microbiology and Immunology
| | - Jen Fossen
- Department of Medicine, Medical Microbiology and Immunology
| | - Hiram Sanchez
- Department of Medicine, Medical Microbiology and Immunology
| | | | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | | | - David R Andes
- Department of Medicine, Medical Microbiology and Immunology
| |
Collapse
|
39
|
Park HS, Price H, Ceballos S, Chi JT, Wax A. Single Cell Analysis of Stored Red Blood Cells Using Ultra-High Throughput Holographic Cytometry. Cells 2021; 10:cells10092455. [PMID: 34572104 PMCID: PMC8465484 DOI: 10.3390/cells10092455] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 01/05/2023] Open
Abstract
Holographic cytometry is introduced as an ultra-high throughput implementation of quantitative phase imaging of single cells flowing through parallel microfluidic channels. Here, the approach was applied for characterizing the morphology of individual red blood cells during storage under regular blood bank conditions. Samples from five blood donors were examined, over 100,000 cells examined for each, at three time points. The approach allows high-throughput phase imaging of a large number of cells, greatly extending our ability to study cellular phenotypes using individual cell images. Holographic cytology images can provide measurements of multiple physical traits of the cells, including optical volume and area, which are observed to consistently change over the storage time. In addition, the large volume of cell imaging data can serve as training data for machine-learning algorithms. For the study here, logistic regression was used to classify the cells according to the storage time points. The analysis showed that at least 5000 cells are needed to ensure accuracy of the classifiers. Overall, results showed the potential of holographic cytometry as a diagnostic tool.
Collapse
Affiliation(s)
- Han-Sang Park
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; (H.-S.P.); (H.P.); (S.C.)
| | - Hillel Price
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; (H.-S.P.); (H.P.); (S.C.)
| | - Silvia Ceballos
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; (H.-S.P.); (H.P.); (S.C.)
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708, USA;
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Adam Wax
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; (H.-S.P.); (H.P.); (S.C.)
- Correspondence:
| |
Collapse
|
40
|
Rouillard ME, Sutter PA, Durham OR, Willis CM, Crocker SJ. Astrocyte-Derived Extracellular Vesicles (ADEVs): Deciphering their Influences in Aging. Aging Dis 2021; 12:1462-1475. [PMID: 34527422 PMCID: PMC8407882 DOI: 10.14336/ad.2021.0608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are an abundant and dynamic glial cell exclusive to the central nervous system (CNS). In the context of injury, inflammation, and/or diseases of the nervous system, astrocyte responses, termed reactive astrogliosis, are a recognized pathological feature across a range of conditions and diseases. However, the impact of reactive astrogliosis is not uniform and varies by context and duration (time). In recent years, extracellular communication between glial cells via extracellular vesicles (EVs) has garnered interest as a process connected with reactive astrogliosis. In this review, we relate recent findings on astrocyte-derived extracellular vesicles (ADEVs) with a focus on factors that can influence the effects of ADEVs and identified age related changes in the function of ADEVs. Additionally, we will discuss the current limitations of existing experimental approaches and identify questions that highlight areas for growth in this field, which will continue to enhance our understanding of ADEVs in age-associated processes.
Collapse
Affiliation(s)
- Megan E Rouillard
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Pearl A Sutter
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Olivia R Durham
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Cory M Willis
- 2Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Stephen J Crocker
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| |
Collapse
|
41
|
Grangier A, Branchu J, Volatron J, Piffoux M, Gazeau F, Wilhelm C, Silva AKA. Technological advances towards extracellular vesicles mass production. Adv Drug Deliv Rev 2021; 176:113843. [PMID: 34147532 DOI: 10.1016/j.addr.2021.113843] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are becoming essential actors in bio-therapeutics, as much for their regenerative or immunomodulatory properties as for their potential as cargo delivery vehicles. To enable the democratization of these EV-based therapies, many challenges remain such as large-scale production which is necessary to reduce costs of treatment. Herein, we review some advanced works on high-yield EV manufacturing. One approach consists in developing large-scale cell culture platforms, while others focus on cell stimulation to increase particle yield per cell. This can be done by moderate physico-chemical stresses or by disrupting cell membrane towards autoassembled vesicle-like particles. We critically compare these different techniques, keeping in mind that the field still lacks shared characterization standards, underline the importance of therapeutic potency assessment and discuss mass production strategies that have been identified in current clinical trials.
Collapse
Affiliation(s)
- Alice Grangier
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France
| | | | | | - Max Piffoux
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France; Everzom, 75006 Paris, France; Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Florence Gazeau
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France
| | - Claire Wilhelm
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France; Laboratoire PhysicoChimie Curie, Institut Curie, PSL Research University - Sorbonne Université - CNRS, 75005 Paris, France.
| | - Amanda K A Silva
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France.
| |
Collapse
|
42
|
Desideri E, Ciccarone F, Ciriolo MR, Fratantonio D. Extracellular vesicles in endothelial cells: from mediators of cell-to-cell communication to cargo delivery tools. Free Radic Biol Med 2021; 172:508-520. [PMID: 34214634 DOI: 10.1016/j.freeradbiomed.2021.06.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles released from most cell types that play a key role in cell-to-cell communication by carrying DNA, non-coding RNAs, proteins and lipids out of cells. The composition of EVs depends on the cell or tissue of origin and changes according to their pathophysiological conditions, making EVs a potential circulating biomarker of disease. Additionally, the natural tropism of EVs for specific organs and cells has raised the interest in their use as delivery vehicles. In this review, we provide an overview of EV biogenesis, isolation and characterization. We also discuss EVs in the context of endothelial pathophysiology, summarizing the current knowledge about their role in cell communication in quiescent and activated endothelial cells. In the last part, we describe the potential use of EVs as delivery vehicles of bioactive compounds and the current strategies to load exogenous cargo and to functionalize EVs to drive them to a specific tissue.
Collapse
Affiliation(s)
- Enrico Desideri
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome
| | - Fabio Ciccarone
- IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome; IRCCS San Raffaele Pisana, Via della Pisana 235, 00163, Rome, Italy.
| | - Deborah Fratantonio
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, 70125 Bari, Italy.
| |
Collapse
|
43
|
Bates M, Watts S, Doughty H, Woolley T, Miles A, Barry L, Jenner D, Sedman A, Purcell R, Kirkman E. Effect of parachute delivery on red blood cell (RBC) and plasma quality measures of blood for transfusion. Transfusion 2021; 61 Suppl 1:S223-S233. [PMID: 34269457 PMCID: PMC9290599 DOI: 10.1111/trf.16501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 11/29/2022]
Abstract
Background Parachute airdrop offers a rapid transfusion supply option for humanitarian aid and military support. However, its impact on longer‐term RBC survival is undocumented. This study aimed to determine post‐drop quality of RBCs in concentrates (RCC), and both RBCs and plasma in whole blood (WB) during subsequent storage. Study design and methods Twenty‐two units of leucodepleted RCC in saline, adenine, glucose, mannitol (SAGM) and 22 units of nonclinical issue WB were randomly allocated for air transportation, parachute drop, and subsequent storage (parachute), or simply storage under identical conventional conditions (4 ± 2°C) (control). All blood products were 6–8 days post‐donation. Parachute units were packed into Credo Cubes, (Series 4, 16 L) inside a PeliCase (Peli 0350) and rigged as parachute delivery packs. Packs underwent a 4‐h tactical flight (C130 aircraft), then parachuted from 250 to 400 ft before ground recovery. The units were sampled aseptically before and after airdrop at weekly intervals. A range of assays quantified the RBC storage lesion and coagulation parameters. Results Blood units were maintained at 2–6°C and recovered intact after recorded ground impacts of 341–1038 m s−2. All units showed a classical RBC storage lesion and increased RBC microparticles during 42 days of storage. Fibrinogen and clotting factors decreased in WB during storage. Nevertheless, no significant difference was observed between Control and Parachute groups. Air transportation and parachute delivery onto land did not adversely affect, or shorten, the shelf life of fresh RBCs or WB. Discussion Appropriately packaged aerial delivery by parachute can be successfully used for blood supply.
Collapse
Affiliation(s)
- Mark Bates
- CBR Division, Dstl Porton Down, Salisbury, UK
| | - Sarah Watts
- CBR Division, Dstl Porton Down, Salisbury, UK
| | - Heidi Doughty
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Translational Medicine, Birmingham, UK.,NHS Blood and Transplant, Birmingham, UK
| | - Tom Woolley
- Academic Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, Birmingham, UK
| | - Andrew Miles
- Centre of Defence Pathology, Royal Centre for Defence Medicine, Birmingham, UK
| | - Liam Barry
- Centre of Defence Pathology, Royal Centre for Defence Medicine, Birmingham, UK
| | | | - Andrew Sedman
- Platform Systems Division, Dstl Porton Down, Salisbury, UK
| | | | | |
Collapse
|
44
|
RoŽanc J, Finšgar M, Maver U. Progressive use of multispectral imaging flow cytometry in various research areas. Analyst 2021; 146:4985-5007. [PMID: 34337638 DOI: 10.1039/d1an00788b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multi-spectral imaging flow cytometry (MIFC) has become one of the most powerful technologies for investigating general analytics, molecular and cell biology, biotechnology, medicine, and related fields. It combines the capabilities of the morphometric and photometric analysis of single cells and micrometer-sized particles in flux with regard to thousands of events. It has become the tool of choice for a wide range of research and clinical applications. By combining the features of flow cytometry and fluorescence microscopy, it offers researchers the ability to couple the spatial resolution of multicolour images of cells and organelles with the simultaneous analysis of a large number of events in a single system. This provides the opportunity to visually confirm findings and collect novel data that would otherwise be more difficult to obtain. This has led many researchers to design innovative assays to gain new insight into important research questions. To date, it has been successfully used to study cell morphology, surface and nuclear protein co-localization, protein-protein interactions, cell signaling, cell cycle, cell death, and cytotoxicity, intracellular calcium, drug uptake, pathogen internalization, and other applications. Herein we describe some of the recent advances in the field of multiparametric imaging flow cytometry methods in various research areas.
Collapse
Affiliation(s)
- Jan RoŽanc
- University of Maribor, Faculty of Medicine, Institute of Biomedical Sciences, Taborska ulica 8, SI-2000 Maribor, Slovenia.
| | | | | |
Collapse
|
45
|
Erdbrügger U, Blijdorp CJ, Bijnsdorp IV, Borràs FE, Burger D, Bussolati B, Byrd JB, Clayton A, Dear JW, Falcón‐Pérez JM, Grange C, Hill AF, Holthöfer H, Hoorn EJ, Jenster G, Jimenez CR, Junker K, Klein J, Knepper MA, Koritzinsky EH, Luther JM, Lenassi M, Leivo J, Mertens I, Musante L, Oeyen E, Puhka M, van Royen ME, Sánchez C, Soekmadji C, Thongboonkerd V, van Steijn V, Verhaegh G, Webber JP, Witwer K, Yuen PS, Zheng L, Llorente A, Martens‐Uzunova ES. Urinary extracellular vesicles: A position paper by the Urine Task Force of the International Society for Extracellular Vesicles. J Extracell Vesicles 2021; 10:e12093. [PMID: 34035881 PMCID: PMC8138533 DOI: 10.1002/jev2.12093] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
Urine is commonly used for clinical diagnosis and biomedical research. The discovery of extracellular vesicles (EV) in urine opened a new fast-growing scientific field. In the last decade urinary extracellular vesicles (uEVs) were shown to mirror molecular processes as well as physiological and pathological conditions in kidney, urothelial and prostate tissue. Therefore, several methods to isolate and characterize uEVs have been developed. However, methodological aspects of EV separation and analysis, including normalization of results, need further optimization and standardization to foster scientific advances in uEV research and a subsequent successful translation into clinical practice. This position paper is written by the Urine Task Force of the Rigor and Standardization Subcommittee of ISEV consisting of nephrologists, urologists, cardiologists and biologists with active experience in uEV research. Our aim is to present the state of the art and identify challenges and gaps in current uEV-based analyses for clinical applications. Finally, recommendations for improved rigor, reproducibility and interoperability in uEV research are provided in order to facilitate advances in the field.
Collapse
|
46
|
Oggero S, de Gaetano M, Marcone S, Fitzsimons S, Pinto AL, Ikramova D, Barry M, Burke D, Montero-Melendez T, Cooper D, Burgoyne T, Belton O, Norling LV, Brennan EP, Godson C, Perretti M. Extracellular vesicles from monocyte/platelet aggregates modulate human atherosclerotic plaque reactivity. J Extracell Vesicles 2021; 10:12084. [PMID: 33936566 PMCID: PMC8077084 DOI: 10.1002/jev2.12084] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging as key players in different stages of atherosclerosis. Here we provide evidence that EVs released by mixed aggregates of monocytes and platelets in response to TNF‐α display pro‐inflammatory actions on endothelial cells and atherosclerotic plaques. Tempering platelet activation with Iloprost, Aspirin or a P2Y12 inhibitor impacted quantity and phenotype of EV produced. Proteomics of EVs from cells activated with TNF‐α alone or in the presence of Iloprost revealed a distinct composition, with interesting hits like annexin‐A1 and gelsolin. When added to human atherosclerotic plaque explants, EVs from TNF‐α stimulated monocytes augmented release of cytokines. In contrast, EVs generated by TNF‐α together with Iloprost produced minimal plaque activation. Notably, patients with coronary artery disease that required percutaneous coronary intervention had elevated plasma numbers of monocyte, platelet as well as double positive EV subsets. In conclusion, EVs released following monocyte/platelet activation may play a potential role in the development and progression of atherosclerosis. Whereas attenuating platelet activation modifies EV composition released from monocyte/platelet aggregates, curbing their pro‐inflammatory actions may offer therapeutic avenues for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Silvia Oggero
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK
| | - Monica de Gaetano
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Simone Marcone
- Trinity Translational Medicine Institute Trinity College Dublin Dublin Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | | | - Dinara Ikramova
- School of Engineering and Materials Science Queen Mary University of London London UK
| | - Mary Barry
- Department of Vascular Surgery St. Vincent's University Hospital Dublin Ireland
| | - David Burke
- Department of Vascular Surgery St. Vincent's University Hospital Dublin Ireland
| | - Trinidad Montero-Melendez
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Dianne Cooper
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Thomas Burgoyne
- Royal Brompton & Harefield NHS Foundation Trust London UK.,Institute of Ophthalmology, Faculty of Brain Sciences University College London London UK
| | - Orina Belton
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Lucy V Norling
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Eoin P Brennan
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Mauro Perretti
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| |
Collapse
|
47
|
Adams SD, Csere J, D'angelo G, Carter EP, Romao M, Arnandis T, Dodel M, Kocher HM, Grose R, Raposo G, Mardakheh F, Godinho SA. Centrosome amplification mediates small extracellular vesicle secretion via lysosome disruption. Curr Biol 2021; 31:1403-1416.e7. [PMID: 33592190 PMCID: PMC8047808 DOI: 10.1016/j.cub.2021.01.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/01/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023]
Abstract
Bidirectional communication between cells and their surrounding environment is critical in both normal and pathological settings. Extracellular vesicles (EVs), which facilitate the horizontal transfer of molecules between cells, are recognized as an important constituent of cell-cell communication. In cancer, alterations in EV secretion contribute to the growth and metastasis of tumor cells. However, the mechanisms underlying these changes remain largely unknown. Here, we show that centrosome amplification is associated with and sufficient to promote small extracellular vesicle (SEV) secretion in pancreatic cancer cells. This is a direct result of lysosomal dysfunction, caused by increased reactive oxygen species (ROS) downstream of extra centrosomes. We propose that defects in lysosome function could promote multivesicular body fusion with the plasma membrane, thereby enhancing SEV secretion. Furthermore, we find that SEVs secreted in response to amplified centrosomes are functionally distinct and activate pancreatic stellate cells (PSCs). These activated PSCs promote the invasion of pancreatic cancer cells in heterotypic 3D cultures. We propose that SEVs secreted by cancer cells with amplified centrosomes influence the bidirectional communication between the tumor cells and the surrounding stroma to promote malignancy.
Collapse
Affiliation(s)
- Sophie D Adams
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Judit Csere
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gisela D'angelo
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Maryse Romao
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Teresa Arnandis
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Martin Dodel
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Richard Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Graça Raposo
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Faraz Mardakheh
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Susana A Godinho
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
48
|
Marki A, Buscher K, Lorenzini C, Meyer M, Saigusa R, Fan Z, Yeh YT, Hartmann N, Dan JM, Kiosses WB, Golden GJ, Ganesan R, Winkels H, Orecchioni M, McArdle S, Mikulski Z, Altman Y, Bui J, Kronenberg M, Chien S, Esko JD, Nizet V, Smalley D, Roth J, Ley K. Elongated neutrophil-derived structures are blood-borne microparticles formed by rolling neutrophils during sepsis. J Exp Med 2021; 218:e20200551. [PMID: 33275138 PMCID: PMC7721910 DOI: 10.1084/jem.20200551] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/28/2020] [Accepted: 11/06/2020] [Indexed: 12/30/2022] Open
Abstract
Rolling neutrophils form tethers with submicron diameters. Here, we report that these tethers detach, forming elongated neutrophil-derived structures (ENDS) in the vessel lumen. We studied ENDS formation in mice and humans in vitro and in vivo. ENDS do not contain mitochondria, endoplasmic reticulum, or DNA, but are enriched for S100A8, S100A9, and 57 other proteins. Within hours of formation, ENDS round up, and some of them begin to present phosphatidylserine on their surface (detected by annexin-5 binding) and release S100A8-S100A9 complex, a damage-associated molecular pattern protein that is a known biomarker of neutrophilic inflammation. ENDS appear in blood plasma of mice upon induction of septic shock. Compared with healthy donors, ENDS are 10-100-fold elevated in blood plasma of septic patients. Unlike neutrophil-derived extracellular vesicles, most ENDS are negative for the tetraspanins CD9, CD63, and CD81. We conclude that ENDS are a new class of bloodborne submicron particles with a formation mechanism linked to neutrophil rolling on the vessel wall.
Collapse
Affiliation(s)
- Alex Marki
- La Jolla Institute for Immunology, La Jolla, CA
| | - Konrad Buscher
- La Jolla Institute for Immunology, La Jolla, CA
- Division of General Internal Medicine, Nephrology, and Rheumatology, Department of Medicine D, University Hospital Muenster, Muenster, Germany
| | - Cristina Lorenzini
- La Jolla Institute for Immunology, La Jolla, CA
- Laboratory of Immunobiology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | | | | | - Zhichao Fan
- La Jolla Institute for Immunology, La Jolla, CA
- Department of Immunology, University of Connecticut Health Center, Farmington, CT
| | - Yi-Ting Yeh
- Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA
| | | | - Jennifer M. Dan
- La Jolla Institute for Immunology, La Jolla, CA
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA
| | | | - Gregory J. Golden
- Department of Cellular and Molecular Medicine and Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Rajee Ganesan
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | - Yoav Altman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Jack Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | | | - Shu Chien
- Institute for Immunology, University of Muenster, Muenster, Germany
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine and Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA
| | - David Smalley
- Systems Mass Spectrometry Core, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Johannes Roth
- Institute for Immunology, University of Muenster, Muenster, Germany
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
49
|
Varrone E, Carnicelli D, Brigotti M. Extracellular Vesicles and Renal Endothelial Cells: A Fatal Attraction in Hemolytic Uremic Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:795-804. [PMID: 33652019 DOI: 10.1016/j.ajpath.2021.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/22/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022]
Abstract
This review focuses on typical hemolytic uremic syndrome (HUS), a life-threatening sequela of human infections caused, particularly in children, by Shiga toxin-producing Escherichia coli strains. Thrombotic microangiopathy of the brain and the kidney is the end point of toxin action, resulting in the hallmarks of HUS (ie, thrombocytopenia, anemia, and acute renal failure). A growing body of evidence points to the role of extracellular vesicles released in the blood of patients by toxin-challenged circulating cells (monocytes, neutrophils, and erythrocytes) and platelets, as a key factor in the pathogenesis of HUS. This review provides i) an updated description of the pathogenesis of Shiga toxin-producing E. coli infections; ii) an analysis of blood cell-derived extracellular vesicles, and of their parent cells, as triggering factors in HUS; and iii) a model explaining why Shiga toxin-containing vesicles dock preferentially to the endothelia of target organs.
Collapse
Affiliation(s)
- Elisa Varrone
- Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Bologna, Italy
| | - Domenica Carnicelli
- Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Bologna, Italy
| | - Maurizio Brigotti
- Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Bologna, Italy.
| |
Collapse
|
50
|
Nederveen JP, Warnier G, Di Carlo A, Nilsson MI, Tarnopolsky MA. Extracellular Vesicles and Exosomes: Insights From Exercise Science. Front Physiol 2021; 11:604274. [PMID: 33597890 PMCID: PMC7882633 DOI: 10.3389/fphys.2020.604274] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
The benefits of exercise on health and longevity are well-established, and evidence suggests that these effects are partially driven by a spectrum of bioactive molecules released into circulation during exercise (e.g., exercise factors or 'exerkines'). Recently, extracellular vesicles (EVs), including microvesicles (MVs) and exosomes or exosome-like vesicles (ELVs), were shown to be secreted concomitantly with exerkines. These EVs have therefore been proposed to act as cargo carriers or 'mediators' of intercellular communication. Given these findings, there has been a rapidly growing interest in the role of EVs in the multi-systemic, adaptive response to exercise. This review aims to summarize our current understanding of the effects of exercise on MVs and ELVs, examine their role in the exercise response and long-term adaptations, and highlight the main methodological hurdles related to blood collection, purification, and characterization of ELVs.
Collapse
Affiliation(s)
- Joshua P Nederveen
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Geoffrey Warnier
- Institut of Neuroscience, UCLouvain, Université catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Alessia Di Carlo
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mats I Nilsson
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada.,Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| |
Collapse
|