51
|
Rodriguez BV, Wen Y, Shirk EN, Vazquez S, Gololobova O, Maxwell A, Plunkard J, Castell N, Carlson B, Queen SE, Izzi JM, Driedonks TAP, Witwer KW. An ex vivo model of interactions between extracellular vesicles and peripheral mononuclear blood cells in whole blood. J Extracell Vesicles 2023; 12:e12368. [PMID: 38047476 PMCID: PMC10694845 DOI: 10.1002/jev2.12368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Accepted: 09/13/2023] [Indexed: 12/05/2023] Open
Abstract
Extracellular vesicles (EVs) can be loaded with therapeutic cargo and engineered for retention by specific body sites; therefore, they have great potential for targeted delivery of biomolecules to treat diseases. However, the pharmacokinetics and biodistribution of EVs in large animals remain relatively unknown, especially in primates. We recently reported that when cell culture-derived EVs are administered intravenously to Macaca nemestrina (pig-tailed macaques), they differentially associate with specific subsets of peripheral blood mononuclear cells (PBMCs). More than 60% of CD20+ B cells were observed to associate with EVs for up to 1 h post-intravenous administration. To investigate these associations further, we developed an ex vivo model of whole blood collected from healthy pig-tailed macaques. Using this ex vivo system, we found that labelled EVs preferentially associate with B cells in whole blood at levels similar to those detected in vivo. This study demonstrates that ex vivo blood can be used to study EV-blood cell interactions.
Collapse
Affiliation(s)
- Blanca V. Rodriguez
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Yi Wen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Erin N. Shirk
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Samuel Vazquez
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Amanda Maxwell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jessica Plunkard
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Tom A. P. Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
52
|
Honda A, Nozumi M, Ito Y, Natsume R, Kawasaki A, Nakatsu F, Abe M, Uchino H, Matsushita N, Ikeda K, Arita M, Sakimura K, Igarashi M. Very-long-chain fatty acids are crucial to neuronal polarity by providing sphingolipids to lipid rafts. Cell Rep 2023; 42:113195. [PMID: 37816355 DOI: 10.1016/j.celrep.2023.113195] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/19/2023] [Accepted: 09/14/2023] [Indexed: 10/12/2023] Open
Abstract
Fatty acids have long been considered essential to brain development; however, the involvement of their synthesis in nervous system formation is unclear. We generate mice with knockout of GPSN2, an enzyme for synthesis of very-long-chain fatty acids (VLCFAs) and investigate the effects. Both GPSN2-/- and GPSN2+/- mice show abnormal neuronal networks as a result of impaired neuronal polarity determination. Lipidomics of GPSN2-/- embryos reveal that ceramide synthesis is specifically inhibited depending on FA length; namely, VLCFA-containing ceramide is reduced. We demonstrate that lipid rafts are highly enriched in growth cones and that GPSN2+/- neurons lose gangliosides in their membranes. Application of C24:0 ceramide, but not C16:0 ceramide or C24:0 phosphatidylcholine, to GPSN2+/- neurons rescues both neuronal polarity determination and lipid-raft density in the growth cone. Taken together, our results indicate that VLCFA synthesis contributes to physiological neuronal development in brain network formation, in particular neuronal polarity determination through the formation of lipid rafts.
Collapse
Affiliation(s)
- Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Center for Research Promotion, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Rie Natsume
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Haruki Uchino
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Natsuki Matsushita
- Division of Laboratory Animal Research, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Kazutaka Ikeda
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
53
|
Xu SQ, Sie ZY, Hsu JI, Tan KT. Small Plasma Membrane-Targeted Fluorescent Dye for Long-Time Imaging and Protein Degradation Analyses. Anal Chem 2023; 95:15549-15555. [PMID: 37816133 DOI: 10.1021/acs.analchem.3c01980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Plasma membrane (PM)-targeted fluorescent dyes have become an important tool to visualize morphological and dynamic changes in the cell membrane. However, most of these PM dyes are either too large and thus might potentially perturb the membrane and affect its functions or exhibit a short retention time on the cell membrane. The rapid internalization problem is particularly severe for PM dyes based on cationic and neutral hydrophobic fluorescent dyes, which can be easily transported into the cells by transmembrane potential and passive diffusion mechanisms. In this paper, we report a small but highly specific PM fluorescent dye, PM-1, which exhibits a very long retention time on the plasma membrane with a half-life of approximately 15 h. For biological applications, we demonstrated that PM-1 can be used in combination with protein labeling probes to study ectodomain shedding and endocytosis processes of cell surface proteins and successfully demonstrated that native transmembrane human carbonic anhydrase IX (hCAIX) is degraded via the ectodomain shedding mechanism. In contrast, hCAIX undergoes endocytic degradation in the presence of sheddase inhibitors. We believe that PM-1 can be a versatile tool to provide detailed insights into the dynamic processes of the cell surface proteins.
Collapse
Affiliation(s)
- Shun-Qiang Xu
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu 30013, Taiwan, Republic of China
| | - Zong-Yan Sie
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu 30013, Taiwan, Republic of China
| | - Jung-I Hsu
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu 30013, Taiwan, Republic of China
| | - Kui-Thong Tan
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu 30013, Taiwan, Republic of China
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, Republic of China
| |
Collapse
|
54
|
Samanta S, Lai K, Wu F, Liu Y, Cai S, Yang X, Qu J, Yang Z. Xanthene, cyanine, oxazine and BODIPY: the four pillars of the fluorophore empire for super-resolution bioimaging. Chem Soc Rev 2023; 52:7197-7261. [PMID: 37743716 DOI: 10.1039/d2cs00905f] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
In the realm of biological research, the invention of super-resolution microscopy (SRM) has enabled the visualization of ultrafine sub-cellular structures and their functions in live cells at the nano-scale level, beyond the diffraction limit, which has opened up a new window for advanced biomedical studies to unravel the complex unknown details of physiological disorders at the sub-cellular level with unprecedented resolution and clarity. However, most of the SRM techniques are highly reliant on the personalized special photophysical features of the fluorophores. In recent times, there has been an unprecedented surge in the development of robust new fluorophore systems with personalized features for various super-resolution imaging techniques. To date, xanthene, cyanine, oxazine and BODIPY cores have been authoritatively utilized as the basic fluorophore units in most of the small-molecule-based organic fluorescent probe designing strategies for SRM owing to their excellent photophysical characteristics and easy synthetic acquiescence. Since the future of next-generation SRM studies will be decided by the availability of advanced fluorescent probes and these four fluorescent building blocks will play an important role in progressive new fluorophore design, there is an urgent need to review the recent advancements in designing fluorophores for different SRM methods based on these fluorescent dye cores. This review article not only includes a comprehensive discussion about the recent developments in designing fluorescent probes for various SRM techniques based on these four important fluorophore building blocks with special emphasis on their effective integration into live cell super-resolution bio-imaging applications but also critically evaluates the background of each of the fluorescent dye cores to highlight their merits and demerits towards developing newer fluorescent probes for SRM.
Collapse
Affiliation(s)
- Soham Samanta
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Kaitao Lai
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Feihu Wu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Yingchao Liu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Songtao Cai
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Xusan Yang
- Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Junle Qu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Zhigang Yang
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
55
|
Tagore M, Hergenreder E, Perlee SC, Cruz NM, Menocal L, Suresh S, Chan E, Baron M, Melendez S, Dave A, Chatila WK, Nsengimana J, Koche RP, Hollmann TJ, Ideker T, Studer L, Schietinger A, White RM. GABA Regulates Electrical Activity and Tumor Initiation in Melanoma. Cancer Discov 2023; 13:2270-2291. [PMID: 37553760 PMCID: PMC10551668 DOI: 10.1158/2159-8290.cd-23-0389] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/27/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Oncogenes can initiate tumors only in certain cellular contexts, which is referred to as oncogenic competence. In melanoma, whether cells in the microenvironment can endow such competence remains unclear. Using a combination of zebrafish transgenesis coupled with human tissues, we demonstrate that GABAergic signaling between keratinocytes and melanocytes promotes melanoma initiation by BRAFV600E. GABA is synthesized in melanoma cells, which then acts on GABA-A receptors in keratinocytes. Electron microscopy demonstrates specialized cell-cell junctions between keratinocytes and melanoma cells, and multielectrode array analysis shows that GABA acts to inhibit electrical activity in melanoma/keratinocyte cocultures. Genetic and pharmacologic perturbation of GABA synthesis abrogates melanoma initiation in vivo. These data suggest that GABAergic signaling across the skin microenvironment regulates the ability of oncogenes to initiate melanoma. SIGNIFICANCE This study shows evidence of GABA-mediated regulation of electrical activity between melanoma cells and keratinocytes, providing a new mechanism by which the microenvironment promotes tumor initiation. This provides insights into the role of the skin microenvironment in early melanomas while identifying GABA as a potential therapeutic target in melanoma. See related commentary by Ceol, p. 2128. This article is featured in Selected Articles from This Issue, p. 2109.
Collapse
Affiliation(s)
- Mohita Tagore
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emiliano Hergenreder
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, New York
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, New York
- Weill Graduate School of Medical Sciences of Cornell University, New York, New York
| | - Sarah C. Perlee
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nelly M. Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura Menocal
- Weill Graduate School of Medical Sciences of Cornell University, New York, New York
| | - Shruthy Suresh
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric Chan
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maayan Baron
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, California
| | - Stephanie Melendez
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Walid K. Chatila
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Richard P. Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Travis J. Hollmann
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, California
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, New York
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard M. White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
56
|
Musicò A, Zenatelli R, Romano M, Zendrini A, Alacqua S, Tassoni S, Paolini L, Urbinati C, Rusnati M, Bergese P, Pomarico G, Radeghieri A. Surface functionalization of extracellular vesicle nanoparticles with antibodies: a first study on the protein corona "variable". NANOSCALE ADVANCES 2023; 5:4703-4717. [PMID: 37705771 PMCID: PMC10496878 DOI: 10.1039/d3na00280b] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/19/2023] [Indexed: 09/15/2023]
Abstract
To be profitably exploited in medicine, nanosized systems must be endowed with biocompatibility, targeting capability, the ability to evade the immune system, and resistance to clearance. Currently, biogenic nanoparticles, such as extracellular vesicles (EVs), are intensively investigated as the platform that naturally recapitulates these highly needed characteristics. EV native targeting properties and pharmacokinetics can be further augmented by decorating the EV surface with specific target ligands as antibodies. However, to date, studies dealing with the functionalization of the EV surface with proteins have never considered the protein corona "variable", namely the fact that extrinsic proteins may spontaneously adsorb on the EV surface, contributing to determine the surface, and in turn the biological identity of the EV. In this work, we explore and compare the two edge cases of EVs modified with the antibody Cetuximab (CTX) by chemisorption of CTX (through covalent binding via biorthogonal click-chemistry) and by formation of a physisorbed CTX corona. The results indicate that (i) no differences exist between the two formulations in terms of binding affinity imparted by molecular recognition of CTX versus its natural binding partner (epidermal growth factor receptor, EGFR), but (ii) significant differences emerge at the cellular level, where CTX-EVs prepared by click chemistry display superior binding and uptake toward target cells, very likely due to the higher robustness of the CTX anchorage.
Collapse
Affiliation(s)
- Angelo Musicò
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Rossella Zenatelli
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Miriam Romano
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Andrea Zendrini
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Silvia Alacqua
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Selene Tassoni
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
| | - Lucia Paolini
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia 25123 Brescia Italy
| | - Chiara Urbinati
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
- National Center for Gene Therapy and Drugs Based on RNA Technology - CN3 Padova Italy
| | - Giuseppe Pomarico
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia Viale Europa 11 25123 Brescia Italy
- CSGI, Center for Colloid and Surface Science 50019 Florence Italy
| |
Collapse
|
57
|
Bao C, Xiang H, Chen Q, Zhao Y, Gao Q, Huang F, Mao L. A Review of Labeling Approaches Used in Small Extracellular Vesicles Tracing and Imaging. Int J Nanomedicine 2023; 18:4567-4588. [PMID: 37588627 PMCID: PMC10426735 DOI: 10.2147/ijn.s416131] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023] Open
Abstract
Small extracellular vesicles (sEVs), a subset of extracellular vesicles (EVs) originating from the endosomal compartment, are a kind of lipid bilayer vesicles released by almost all types of cells, serving as natural carriers of nucleic acids, proteins, and lipids for intercellular communication and transfer of bioactive molecules. The current findings suggest their vital role in physiological and pathological processes. Various sEVs labeling techniques have been developed for the more advanced study of the function, mode of action, bio-distribution, and related information of sEVs. In this review, we summarize the existing and emerging sEVs labeling techniques, including fluorescent labeling, radioisotope labeling, nanoparticle labeling, chemical contrast agents labeling, and label-free technique. These approaches will pave the way for an in-depth study of sEVs. We present a systematic and comprehensive review of the principles, advantages, disadvantages, and applications of these techniques, to help promote applications of these labeling approaches in future research on sEVs.
Collapse
Affiliation(s)
- Chenxuan Bao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Huayuan Xiang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Qiaoqiao Chen
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Yuxue Zhao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Qianqian Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Feng Huang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Lingxiang Mao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| |
Collapse
|
58
|
Sternstein C, Böhm TM, Fink J, Meyr J, Lüdemann M, Krug M, Kriukov K, Gurdap CO, Sezgin E, Ebert R, Seibel J. Development of an Effective Functional Lipid Anchor for Membranes (FLAME) for the Bioorthogonal Modification of the Lipid Bilayer of Mesenchymal Stromal Cells. Bioconjug Chem 2023; 34:1221-1233. [PMID: 37328799 DOI: 10.1021/acs.bioconjchem.3c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The glycosylation of cellular membranes is crucial for the survival and communication of cells. As our target is the engineering of the glycocalyx, we designed a functionalized lipid anchor for the introduction into cellular membranes called Functional Lipid Anchor for MEmbranes (FLAME). Since cholesterol incorporates very effectively into membranes, we developed a twice cholesterol-substituted anchor in a total synthesis by applying protecting group chemistry. We labeled the compound with a fluorescent dye, which allows cell visualization. FLAME was successfully incorporated in the membranes of living human mesenchymal stromal cells (hMSC), acting as a temporary, nontoxic marker. The availability of an azido function─a bioorthogonal reacting group within the compound─enables the convenient coupling of alkyne-functionalized molecules, such as fluorophores or saccharides. After the incorporation of FLAME into the plasma membrane of living hMSC, we were able to successfully couple our molecule with an alkyne-tagged fluorophore via click reaction. This suggests that FLAME is useful for the modification of the membrane surface. Coupling FLAME with a galactosamine derivative yielded FLAME-GalNAc, which was incorporated into U2OS cells as well as in giant unilamellar vesicles (GUVs) and cell-derived giant plasma membrane vesicles (GPMVs). With this, we have shown that FLAME-GalNAc is a useful tool for studying the partitioning in the liquid-ordered (Lo) and the liquid-disordered (Ld) phases. The molecular tool can also be used to analyze the diffusion behavior in the model and the cell membranes by fluorescence correlation spectroscopy (FCS).
Collapse
Affiliation(s)
- Christine Sternstein
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Theresa-Maria Böhm
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Julian Fink
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Jessica Meyr
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Martin Lüdemann
- Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Würzburg, Brettreichstr. 11, 97074 Würzburg, Germany
| | - Melanie Krug
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Kirill Kriukov
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Cenk O Gurdap
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Regina Ebert
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
59
|
Abstract
A wide range of biomaterials and engineered cell surfaces are composed of bioconjugates embedded in liposome membranes, surface-immobilized bilayers, or the plasma membranes of living cells. This review article summarizes the various ways that Nature anchors integral and peripheral proteins in a cell membrane and describes the strategies devised by chemical biologists to label a membrane protein in living cells. Also discussed are modern synthetic and semisynthetic methods to produce lipidated proteins. Subsequent sections describe methods to anchor a three-component synthetic construct that is composed of a lipophilic membrane anchor, hydrophilic linker, and exposed functional component. The surface exposed payload can be a fluorophore, aptamer, oligonucleotide, polypeptide, peptide nucleic acid, polysaccharide, branched dendrimer, or linear polymer. Hydrocarbon chains are commonly used as the membrane anchor, and a general experimental trend is that a two chain lipid anchor has higher membrane affinity than a cholesteryl or single chain lipid anchor. Amphiphilic fluorescent dyes are effective molecular probes for cell membrane imaging and a zwitterionic linker between the fluorophore and the lipid anchor promotes high persistence in the plasma membrane of living cells. A relatively new advance is the development of switchable membrane anchors as molecular tools for fundamental studies or as technology platforms for applied biomaterials.
Collapse
Affiliation(s)
- Rananjaya S Gamage
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jordan L Chasteen
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
60
|
He Y, Xing Y, Jiang T, Wang J, Sang S, Rong H, Yu F. Fluorescence labeling of extracellular vesicles for diverse bio-applications in vitro and in vivo. Chem Commun (Camb) 2023; 59:6609-6626. [PMID: 37161668 DOI: 10.1039/d3cc00998j] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles enclosed in a lipid membrane that are sustainably released by nearly all cell types. EVs have been deemed as valuable biomarkers for diagnostics and effective drug carriers, owing to the physiological function of transporting biomolecules for intercellular communication. To investigate their biological properties, efficient labeling strategies have been constructed for EV research, among which fluorescence labeling exerts a powerful function due to the capability of visualizing the nanovesicles with high sensitivity both in vitro and in vivo. In one aspect, with the help of functional fluorescence tags, EVs could be differentiated and categorized in vitro by various analytical techniques, which exert vital roles in disease diagnosis, prognosis, and treatment monitoring. Additionally, innovative EV reporters have been utilized for visualizing EVs, in combination with powerful microscopy techniques, which provide potential tools for investigating the dynamic events of EV release and intercellular communication in suitable animal models. In this feature article, we survey the latest advances regarding EV fluorescence labeling strategies and their application in biomedical application and in vivo biology investigation, highlighting the progresses in individual EV imaging. Finally, the challenges and future perspectives in unravelling EV physiological properties and further biomedical application are discussed.
Collapse
Affiliation(s)
- Yun He
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Yanlong Xing
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Tongmeng Jiang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Juan Wang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Shenggang Sang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Hong Rong
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Fabiao Yu
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
61
|
Jung SR, Kim J, Vojtech L, Vaughan JC, Chiu DT. Error-Correction Method for High-Throughput Sizing of Nanoscale Vesicles with Single-Molecule Localization Microscopy. J Phys Chem B 2023; 127:2701-2707. [PMID: 36944080 PMCID: PMC10224584 DOI: 10.1021/acs.jpcb.2c09053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Single-molecule localization microscopy (SMLM) allows super-resolution imaging, mapping, counting, and sizing of biological nanostructures such as cell organelles and extracellular vesicles (EVs), but sizing structures smaller than ∼100 nm can be inaccurate due to single-molecule localization error caused by distortion of the point spread function and limited photon number. Here we demonstrate a method to correct localization error when sizing vesicles and other spherical nanoparticles with SMLM and compare sizing results using two vesicle labeling schemes. We use mean approximation theory to derive a simple equation using full width at half-maximum (FWHM) for correcting particle sizes measured by two-dimensional SMLM, validate the method by sizing streptavidin-coated polystyrene nanobeads with the SMLM technique dSTORM with and without error correction, using transmission electron microscopy (TEM) for comparison, and then apply the method to sizing small seminal EVs. Nanobead sizes measured by dSTORM became increasingly less accurate (larger than TEM values) for beads smaller than 50 nm. The error-correction method reduced the size difference versus TEM from 15% without error correction to 7% with error correction for 40 nm beads, from 44% to 9% for 30 nm beads, and from 66% to 15% for 20 nm beads. Seminal EVs were labeled with a lipophilic membrane dye (MemBright 700) and with an Alexa Fluor 488-anti-CD63 antibody conjugate, and were sized separately using both dyes by dSTORM. Error-corrected exosome diameters were smaller than uncorrected values: 72 nm vs 79 nm mean diameter with membrane dyes; 84 nm vs 97 nm with the antibody-conjugated dyes. The mean error-corrected diameter was 12 nm smaller when using the membrane dye than when using the antibody-conjugated dye likely due to the large size of the antibody. Thus, both the error-correction method and the compact membrane labeling scheme reduce overestimation of vesicle size by SMLM. This error-correction method has a low computational cost as it does not require correction of individual blinking events, and it is compatible with all SMLM techniques (e.g., PALM, STORM, and DNA-PAINT).
Collapse
Affiliation(s)
- Seung-Ryoung Jung
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - James Kim
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Lucia Vojtech
- Departments of Obstetrics and Gynecology,University of Washington, Seattle, WA 98195, USA
| | - Joshua C. Vaughan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
- Departments of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Daniel T. Chiu
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
62
|
Serrano-Buitrago S, Muñoz-Úbeda M, Almendro-Vedia VG, Sánchez-Camacho J, Maroto BL, Moreno F, Bañuelos J, García-Moreno I, López-Montero I, de la Moya S. Polar ammoniostyryls easily converting a clickable lipophilic BODIPY in an advanced plasma membrane probe. J Mater Chem B 2023; 11:2108-2114. [PMID: 36808432 DOI: 10.1039/d2tb02516g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
A very simple, small and symmetric, but highly bright, photostable and functionalizable molecular probe for plasma membrane (PM) has been developed from an accessible, lipophilic and clickable organic dye based on BODIPY. To this aim, two lateral polar ammoniostyryl groups were easily linked to increase the amphiphilicity of the probe and thus its lipid membrane partitioning. Compared to the BODIPY precursor, the transversal diffusion across lipid bilayers of the ammoniostyryled BODIPY probe was highly reduced, as evidenced by fluorescence confocal microscopy on model membranes built up as giant unilamellar vesicles (GUVs). Moreover, the ammoniostyryl groups endow the new BODIPY probe with the ability to optically work (excitation and emission) in the bioimaging-useful red region, as shown by staining of the plasma membrane of living mouse embryonic fibroblasts (MEFs). Upon incubation, this fluorescent probe rapidly entered the cell through the endosomal pathway. By blocking the endocytic trafficking at 4 °C, the probe was confined within the PM of MEFs. Our experiments show the developed ammoniostyrylated BODIPY as a suitable PM fluorescent probe, and confirm the synthetic approach for advancing PM probes, imaging and science.
Collapse
Affiliation(s)
- Sergio Serrano-Buitrago
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Mónica Muñoz-Úbeda
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain.,Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Víctor G Almendro-Vedia
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain.,Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Juan Sánchez-Camacho
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Beatriz L Maroto
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Florencio Moreno
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Jorge Bañuelos
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco (UPV-EHU), Barrio Sarriena s/n, 48080, Bilbao, Spain
| | - Inmaculada García-Moreno
- Departamento de Química-Física de Materiales, Instituto de Química Física Rocasolano, C.S.I.C., 28006, Madrid, Spain
| | - Iván López-Montero
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain.,Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain. .,Instituto Pluridisciplinar, Universidad Complutense de Madrid, Paseo de Juan XXIII 1, 28040, Madrid, Spain
| | - Santiago de la Moya
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| |
Collapse
|
63
|
Antoniou A, Auderset L, Kaurani L, Sebastian E, Zeng Y, Allahham M, Cases-Cunillera S, Schoch S, Gründemann J, Fischer A, Schneider A. Neuronal extracellular vesicles and associated microRNAs induce circuit connectivity downstream BDNF. Cell Rep 2023; 42:112063. [PMID: 36753414 DOI: 10.1016/j.celrep.2023.112063] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/13/2022] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as mediators of cellular communication, in part via the delivery of associated microRNAs (miRNAs), small non-coding RNAs that regulate gene expression. We show that brain-derived neurotrophic factor (BDNF) mediates the sorting of miR-132-5p, miR-218-5p, and miR-690 in neuron-derived EVs. BDNF-induced EVs in turn increase excitatory synapse formation in recipient hippocampal neurons, which is dependent on the inter-neuronal delivery of these miRNAs. Transcriptomic analysis further indicates the differential expression of developmental and synaptogenesis-related genes by BDNF-induced EVs, many of which are predicted targets of miR-132-5p, miR-218-5p, and miR-690. Furthermore, BDNF-induced EVs up-regulate synaptic vesicle (SV) clustering in a transmissible manner, thereby increasing synaptic transmission and synchronous neuronal activity. As BDNF and EV-miRNAs miR-218 and miR-132 were previously implicated in neuropsychiatric disorders such as anxiety and depression, our results contribute to a better understanding of disorders characterized by aberrant neural circuit connectivity.
Collapse
Affiliation(s)
- Anna Antoniou
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| | - Loic Auderset
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Lalit Kaurani
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany; Department for Systems Medicine and Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, 37075 Göttingen, Germany
| | - Eva Sebastian
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Yuzhou Zeng
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Maria Allahham
- Institute of Bio- and Geosciences 1, Forschungszentrum Jülich, 52428 Jülich, Germany; Aachen Biology and Biotechnology, RWTH Aachen University, 52056 Aachen, Germany
| | - Silvia Cases-Cunillera
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Jan Gründemann
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany; Department for Systems Medicine and Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, 37075 Göttingen, Germany
| | - Anja Schneider
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
64
|
Ohira K, Sato Y, Nishizawa S. Self-Assembly and Disassembly of Membrane Curvature-Sensing Peptide-Based Deep-Red Fluorescent Probe for Highly Sensitive Sensing of Exosomes. ACS Sens 2023; 8:522-526. [PMID: 36695520 DOI: 10.1021/acssensors.2c02498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
With increasing knowledge of the diverse roles of exosomes in biological processes, much attention has been paid to the development of analytical methods for exosome analysis. Here, we developed a new class of amphipathic helical (AH) peptide-based fluorescent probes for highly sensitive detection of exosomes in a mix and read manner. Membrane curvature-sensing AH peptide (ApoC) was coupled with lipophilic tail (C12)-carrying thiazole red (TR) for construction of a self-assembly/disassembly based fluorescence "off-on" sensing system for target exosomes. ApoC-TRC12 has extremely weak emission due to the formation of the aggregates, whereas it becomes emissive in response to the target exosomes through the binding-induced disassembly of ApoC-TRC12. We demonstrated that the C12 unit attached to the TR unit had a favorable effect on both fluorescence response (signal-to-background: S/B) and binding affinity. ApoC-TRC12 was applicable to rapid and simple detection of exosomes with high detection sensitivity (limit of detection ≈ 103 particles/μL).
Collapse
Affiliation(s)
- Kaito Ohira
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki-Aza Aoba, Aoba-ku, Sendai, 980-8578, Japan
| | - Yusuke Sato
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki-Aza Aoba, Aoba-ku, Sendai, 980-8578, Japan.,JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Seiichi Nishizawa
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki-Aza Aoba, Aoba-ku, Sendai, 980-8578, Japan
| |
Collapse
|
65
|
Rzewnicka A, Krysiak J, Pawłowska R, Żurawiński R. Red-Emitting Dithienothiophene S, S-Dioxide Dyes for Cellular Membrane Staining. MATERIALS (BASEL, SWITZERLAND) 2023; 16:ma16051806. [PMID: 36902920 PMCID: PMC10003865 DOI: 10.3390/ma16051806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 06/12/2023]
Abstract
A series of dithienothiophene S,S-dioxide (DTTDO) dyes was designed, synthesized, and investigated for their suitability in fluorescent cell imaging. Synthetized (D-π-A-π-D)-type DTTDO derivatives have molecule lengths close to the thickness of the phospholipid membrane, and they contain on both ends two positively charged or neutral polar groups to increase their solubility in water and to ensure simultaneous interaction with polar groups of the inner and outer part of the cellular membrane. DTTDO derivatives exhibit absorbance and emission maxima in the 517-538 nm and 622-694 nm range, respectively, and a large Stokes shift up to 174 nm. Fluorescence microscopy experiments revealed that these compounds selectively intercalate into cell membranes. Moreover, a cytotoxicity assay conducted on a model human live cells indicates low toxicity of these compounds at the concentrations required for effective staining. With suitable optical properties, low cytotoxicity, and high selectivity against cellular structures, DTTDO derivatives are proven to be attractive dyes for fluorescence-based bioimaging.
Collapse
Affiliation(s)
- Aneta Rzewnicka
- Division of Organic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Jerzy Krysiak
- Division of Organic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Róża Pawłowska
- Division of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Remigiusz Żurawiński
- Division of Organic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
66
|
Xu S, Pan W, Song ZL, Yuan L. Molecular Engineering of Near-Infrared Fluorescent Probes for Cell Membrane Imaging. Molecules 2023; 28:1906. [PMID: 36838896 PMCID: PMC9960866 DOI: 10.3390/molecules28041906] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Cell membrane (CM) is a phospholipid bilayer that maintains integrity of a whole cell and relates to many physiological and pathological processes. Developing CM imaging tools is a feasible method for visualizing membrane-related events. In recent decades, small-molecular fluorescent probes in the near-infrared (NIR) region have been pursued extensively for CM staining to investigate its functions and related events. In this review, we summarize development of such probes from the aspect of design principles, CM-targeting mechanisms and biological applications. Moreover, at the end of this review, the challenges and future research directions in designing NIR CM-targeting probes are discussed. This review indicates that more efforts are required to design activatable NIR CM-targeting probes, easily prepared and biocompatible probes with long retention time regarding CM, super-resolution imaging probes for monitoring CM nanoscale organization and multifunctional probes with imaging and phototherapy effects.
Collapse
Affiliation(s)
- Shuai Xu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Wenjing Pan
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Zhi-Ling Song
- Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Hunan University, Changsha 410082, China
| |
Collapse
|
67
|
Welsh JA, Arkesteijn GJA, Bremer M, Cimorelli M, Dignat-George F, Giebel B, Görgens A, Hendrix A, Kuiper M, Lacroix R, Lannigan J, van Leeuwen TG, Lozano-Andrés E, Rao S, Robert S, de Rond L, Tang VA, Tertel T, Yan X, Wauben MHM, Nolan JP, Jones JC, Nieuwland R, van der Pol E. A compendium of single extracellular vesicle flow cytometry. J Extracell Vesicles 2023; 12:e12299. [PMID: 36759917 PMCID: PMC9911638 DOI: 10.1002/jev2.12299] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 11/29/2022] [Accepted: 12/17/2022] [Indexed: 02/11/2023] Open
Abstract
Flow cytometry (FCM) offers a multiparametric technology capable of characterizing single extracellular vesicles (EVs). However, most flow cytometers are designed to detect cells, which are larger than EVs. Whereas cells exceed the background noise, signals originating from EVs partly overlap with the background noise, thereby making EVs more difficult to detect than cells. This technical mismatch together with complexity of EV-containing fluids causes limitations and challenges with conducting, interpreting and reproducing EV FCM experiments. To address and overcome these challenges, researchers from the International Society for Extracellular Vesicles (ISEV), International Society for Advancement of Cytometry (ISAC), and the International Society on Thrombosis and Haemostasis (ISTH) joined forces and initiated the EV FCM working group. To improve the interpretation, reporting, and reproducibility of future EV FCM data, the EV FCM working group published an ISEV position manuscript outlining a framework of minimum information that should be reported about an FCM experiment on single EVs (MIFlowCyt-EV). However, the framework contains limited background information. Therefore, the goal of this compendium is to provide the background information necessary to design and conduct reproducible EV FCM experiments. This compendium contains background information on EVs, the interaction between light and EVs, FCM hardware, experimental design and preanalytical procedures, sample preparation, assay controls, instrument data acquisition and calibration, EV characterization, and data reporting. Although this compendium focuses on EVs, many concepts and explanations could also be applied to FCM detection of other particles within the EV size range, such as bacteria, lipoprotein particles, milk fat globules, and viruses.
Collapse
Affiliation(s)
- Joshua A Welsh
- Translational Nanobiology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ger J A Arkesteijn
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Michel Bremer
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Cimorelli
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Chemical Engineering, Drexel University, Philadelphia, Pennsylvania, USA
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Clinical Research Center, Department for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Evox Therapeutics Ltd, Oxford, UK
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Martine Kuiper
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Dutch Metrology Institute, VSL, Delft, The Netherlands
| | - Romaric Lacroix
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Joanne Lannigan
- Flow Cytometry Support Services, LLC, Arlington, Virginia, USA
| | - Ton G van Leeuwen
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Estefanía Lozano-Andrés
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Shoaib Rao
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stéphane Robert
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Leonie de Rond
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Vera A Tang
- Flow Cytometry & Virometry Core Facility, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Xiaomei Yan
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Marca H M Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - John P Nolan
- Scintillon Institute, San Diego, California, USA
- Cellarcus Biosciences, San Diego, California, USA
| | - Jennifer C Jones
- Translational Nanobiology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rienk Nieuwland
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Edwin van der Pol
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
68
|
Saladin L, Breton V, Dal Pra O, Klymchenko AS, Danglot L, Didier P, Collot M. Dual-Color Photoconvertible Fluorescent Probes Based on Directed Photooxidation Induced Conversion for Bioimaging. Angew Chem Int Ed Engl 2023; 62:e202215085. [PMID: 36420823 PMCID: PMC10107923 DOI: 10.1002/anie.202215085] [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: 10/14/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
We herein present a new concept to produce dual-color photoconvertible probes based on a mechanism called Directed Photooxidation Induced Conversion (DPIC). As a support of this mechanism, styryl-coumarins (SCs) bearing Aromatic Singlet Oxygen Reactive Moieties (ASORMs) like furan and pyrrole have been synthesized. SCs are bright fluorophores, which undergo a hypsochromic conversion upon visible light irradiation due to directed photooxidation of the ASORM that leads to the disruption of conjugation. SC-P, a yellow emitting probe bearing a pyrrole moiety, converts to a stable blue emitting coumarin with a 68 nm shift allowing the photoconversion and tracking of lipid droplet in live cells. This new approach might pave the way to a new generation of photoconvertible dyes for advanced bioimaging applications.
Collapse
Affiliation(s)
- Lazare Saladin
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/, Université de Strasbourg, 74 route du Rhin, 67401, Illkirch-Graffenstaden, France
| | - Victor Breton
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy and Diseased Brain, Université Paris Cité, 102 rue de la santé, 75014, Paris, France
| | - Ophélie Dal Pra
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/, Université de Strasbourg, 74 route du Rhin, 67401, Illkirch-Graffenstaden, France
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/, Université de Strasbourg, 74 route du Rhin, 67401, Illkirch-Graffenstaden, France
| | - Lydia Danglot
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy and Diseased Brain, Université Paris Cité, 102 rue de la santé, 75014, Paris, France.,Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Sientific director of NeurImag facility, Université Paris Cité, 102 rue de la santé, 75014, Paris, France
| | - Pascal Didier
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/, Université de Strasbourg, 74 route du Rhin, 67401, Illkirch-Graffenstaden, France
| | - Mayeul Collot
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/, Université de Strasbourg, 74 route du Rhin, 67401, Illkirch-Graffenstaden, France
| |
Collapse
|
69
|
Zhou C, Cox-Vázquez SJ, Chia GW, Vázquez RJ, Lai HY, Chan SJ, Limwongyut J, Bazan GC. Water-soluble extracellular vesicle probes based on conjugated oligoelectrolytes. SCIENCE ADVANCES 2023; 9:eade2996. [PMID: 36630497 PMCID: PMC9833659 DOI: 10.1126/sciadv.ade2996] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
We developed a series of transmembrane conjugated oligoelectrolytes (COEs) with tunable optical emissions from the UV to the near IR to address the false-positive problem when detecting nanometer-sized extracellular vesicles (EVs) by flow cytometry. The amphiphilic molecular framework of COEs is defined by a linear conjugated structure and cationic charged groups at each terminal site. Consequently, COEs have excellent water solubility and the absence of nanoaggregates at concentrations up to 50 μM, and unbound COE dyes can be readily removed through ultrafiltration. These properties enable unambiguous and simple detection of COE-labeled small EVs using flow cytometry with negligible background signals. We also demonstrated the time-lapsed tracking of small EV uptake into mammalian cells and the endogenous small EV labeling using COEs. Briefly, COEs provide a class of membrane-targeting dyes that behave as biomimetics of the lipid bilayer and a general and practical labeling strategy for nanosized EVs.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Institute of Polymer Optoelectronic Materials and Devices, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, PR China
| | - Sarah J. Cox-Vázquez
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore 117544, Singapore
| | - Geraldine W. N. Chia
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Ricardo Javier Vázquez
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore 117544, Singapore
| | - Hui Ying Lai
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Samuel J. W. Chan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jakkarin Limwongyut
- Department of Chemistry and Biochemistry, Center for Polymers and Organic Solids, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Guillermo C. Bazan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore 117544, Singapore
- Department of Chemistry and Biochemistry, Center for Polymers and Organic Solids, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| |
Collapse
|
70
|
Quantifying membrane binding and diffusion with fluorescence correlation spectroscopy diffusion laws. Biophys J 2023:S0006-3495(23)00006-1. [PMID: 36632034 DOI: 10.1016/j.bpj.2023.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Many transient processes in cells arise from the binding of cytosolic proteins to membranes. Quantifying this membrane binding and its associated diffusion in the living cell is therefore of primary importance. Dynamic photonic microscopies, e.g., single/multiple particle tracking, fluorescence recovery after photobleaching, and fluorescence correlation spectroscopy (FCS), enable non-invasive measurement of molecular mobility in living cells and their plasma membranes. However, FCS with a single beam waist is of limited applicability with complex, non-Brownian, motions. Recently, the development of FCS diffusion laws methods has given access to the characterization of these complex motions, although none of them is applicable to the membrane binding case at the moment. In this study, we combined computer simulations and FCS experiments to propose an FCS diffusion law for membrane binding. First, we generated computer simulations of spot-variation FCS (svFCS) measurements for a membrane binding process combined to 2D and 3D diffusion at the membrane and in the bulk/cytosol, respectively. Then, using these simulations as a learning set, we derived an empirical diffusion law with three free parameters: the apparent binding constant KD, the diffusion coefficient on the membrane D2D, and the diffusion coefficient in the cytosol, D3D. Finally, we monitored, using svFCS, the dynamics of retroviral Gag proteins and associated mutants during their binding to supported lipid bilayers of different lipid composition or at plasma membranes of living cells, and we quantified KD and D2D in these conditions using our empirical diffusion law. Based on these experiments and numerical simulations, we conclude that this new approach enables correct estimation of membrane partitioning and membrane diffusion properties (KD and D2D) for peripheral membrane molecules.
Collapse
|
71
|
Gargiulo E, Viry E, Morande PE, Largeot A, Gonder S, Xian F, Ioannou N, Benzarti M, Kleine Borgmann FB, Mittelbronn M, Dittmar G, Nazarov PV, Meiser J, Stamatopoulos B, Ramsay AG, Moussay E, Paggetti J. Extracellular Vesicle Secretion by Leukemia Cells In Vivo Promotes CLL Progression by Hampering Antitumor T-cell Responses. Blood Cancer Discov 2023; 4:54-77. [PMID: 36108149 PMCID: PMC9816815 DOI: 10.1158/2643-3230.bcd-22-0029] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/04/2022] [Accepted: 09/07/2022] [Indexed: 01/11/2023] Open
Abstract
Small extracellular vesicle (sEV, or exosome) communication among cells in the tumor microenvironment has been modeled mainly in cell culture, whereas their relevance in cancer pathogenesis and progression in vivo is less characterized. Here we investigated cancer-microenvironment interactions in vivo using mouse models of chronic lymphocytic leukemia (CLL). sEVs isolated directly from CLL tissue were enriched in specific miRNA and immune-checkpoint ligands. Distinct molecular components of tumor-derived sEVs altered CD8+ T-cell transcriptome, proteome, and metabolome, leading to decreased functions and cell exhaustion ex vivo and in vivo. Using antagomiRs and blocking antibodies, we defined specific cargo-mediated alterations on CD8+ T cells. Abrogating sEV biogenesis by Rab27a/b knockout dramatically delayed CLL pathogenesis. This phenotype was rescued by exogenous leukemic sEV or CD8+ T-cell depletion. Finally, high expression of sEV-related genes correlated with poor outcomes in CLL patients, suggesting sEV profiling as a prognostic tool. In conclusion, sEVs shape the immune microenvironment during CLL progression. SIGNIFICANCE sEVs produced in the leukemia microenvironment impair CD8+ T-cell mediated antitumor immune response and are indispensable for leukemia progression in vivo in murine preclinical models. In addition, high expression of sEV-related genes correlated with poor survival and unfavorable clinical parameters in CLL patients. See related commentary by Zhong and Guo, p. 5. This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Ernesto Gargiulo
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Elodie Viry
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Pablo Elías Morande
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg.,Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Anne Largeot
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Susanne Gonder
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Feng Xian
- Proteomics of Cellular Signaling, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Nikolaos Ioannou
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Mohaned Benzarti
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Felix Bruno Kleine Borgmann
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Department of Neurosurgery, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg.,Luxembourg Centre of Neuropathology, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Michel Mittelbronn
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Luxembourg Centre of Neuropathology, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg.,Luxembourg Centre of Neuropathology, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,National Center of Pathology, Laboratoire national de santé (LNS), Dudelange, Luxembourg.,Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Gunnar Dittmar
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Proteomics of Cellular Signaling, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Petr V. Nazarov
- Multiomics Data Science Group, Department of Cancer Research, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Alan G. Ramsay
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Etienne Moussay
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg.,Corresponding Authors: Jérôme Paggetti, Department of Cancer Research, Luxembourg Institute of Health, 6, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg. Phone: 352-26970-344; E-mail: ; and Etienne Moussay. Phone: 352-26970-232; E-mail:
| | - Jérôme Paggetti
- Tumor–Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg.,Corresponding Authors: Jérôme Paggetti, Department of Cancer Research, Luxembourg Institute of Health, 6, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg. Phone: 352-26970-344; E-mail: ; and Etienne Moussay. Phone: 352-26970-232; E-mail:
| |
Collapse
|
72
|
Abstract
Biomembranes are ubiquitous lipid structures that delimit the cell surface and organelles and operate as platforms for a multitude of biomolecular processes. The development of chemical tools─fluorescent probes─for the sensing and imaging of biomembranes is a rapidly growing research direction, stimulated by a high demand from cell biologists and biophysicists. This Account focuses on advances in these smart molecules, providing a voyage from the cell frontier─plasma membranes (PM)─toward intracellular membrane compartments─organelles. General classification of the membrane probes can be based on targeting principles, sensing profile, and optical response. Probes for PM and organelle membranes are designed based on multiple targeting principles: conjugation with natural lipids or synthetic targeting ligands and in situ cell labeling by bio-orthogonal chemistry, conjugation to protein tags, and receptor-ligand interactions. Thus, to obtain membrane probes targeting PM with selectivity to one leaflet, we designed membrane anchor ligands based on a charged group and an alkyl chain. According to the sensing profile, we define basic membrane markers with constant emission and probes for biophysical and chemical sensing. The markers are built from classical fluorophores, exemplified by a series of bright cyanines and BODIPY dyes bearing the PM anchors (MemBright). Membrane probes for biophysical sensing are based on environment-sensitive fluorophores: (1) polarity-sensitive solvatochromic dyes; (2) viscosity-sensitive fluorescent molecular rotors; (3) mechanosensitive fluorescent flippers; and (4) voltage-sensitive electrochromic dyes. Our solvatochromic probes based on Nile Red (NR12S, NR12A, NR4A), Laurdan (Pro12A), and 3-hydroxyflavone (F2N12S) through polarity-sensing can visualize liquid ordered and disordered phases of lipid membranes, sense lipid order and its heterogeneity in cell PM, detect apoptosis, etc. Chemically sensitive probes, combining a dye, membrane-targeting ligand, and molecular recognition unit, enable the detection of pH, ions, redox species, lipids, and proteins at the biomembrane surface. In terms of the optical response profile, we can identify (1) fluorogenic (turn-on) probes, allowing background-free imaging; (2) ratiometric probes, e.g., solvatochromic probes, which enable ratiometric imaging by changing their emission/excitation color; (3) fluorescence lifetime-responsive probes, e.g., fluorescence molecular rotors and flippers, suitable for fluorescence lifetime imaging (FLIM); and (4) switchable probes, important for single-molecule localization microscopy. We showed that combining solvatochromic probes with on-off switching through a reversible binding specifically to cell PM enables the mapping of their biophysical properties with superior resolution. While the majority of efforts have been focused on PM, the probes for cellular organelles, such as endoplasmic reticulum, mitochondria, Golgi apparatus, etc., emerge rapidly. Thus, nontargeted solvatochromic probes can distinguish organelles by the emission color. Targeted solvatochromic probes based on Nile Red revealed unique signatures of polarity and lipid order of individual organelles and their different sensitivities to oxidative or mechanical stress. Lipid droplets, which are membraneless lipidic structures, constitute another interesting organelle target for probing the cell stress. Currently, we stand at the beginning of a long route with big challenges ahead, in particular (1) to achieve superior organelle specificity; (2) to label specific biomembrane leaflets, notably the inner leaflet of PM; (3) to detect lipid organization in a proximity of specific proteins; and (4) to probe biomembranes in tissues and animals.
Collapse
Affiliation(s)
- Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| |
Collapse
|
73
|
Kikuchi K, Adair LD, Lin J, New EJ, Kaur A. Photochemical Mechanisms of Fluorophores Employed in Single-Molecule Localization Microscopy. Angew Chem Int Ed Engl 2023; 62:e202204745. [PMID: 36177530 PMCID: PMC10100239 DOI: 10.1002/anie.202204745] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Indexed: 02/02/2023]
Abstract
Decoding cellular processes requires visualization of the spatial distribution and dynamic interactions of biomolecules. It is therefore not surprising that innovations in imaging technologies have facilitated advances in biomedical research. The advent of super-resolution imaging technologies has empowered biomedical researchers with the ability to answer long-standing questions about cellular processes at an entirely new level. Fluorescent probes greatly enhance the specificity and resolution of super-resolution imaging experiments. Here, we introduce key super-resolution imaging technologies, with a brief discussion on single-molecule localization microscopy (SMLM). We evaluate the chemistry and photochemical mechanisms of fluorescent probes employed in SMLM. This Review provides guidance on the identification and adoption of fluorescent probes in single molecule localization microscopy to inspire the design of next-generation fluorescent probes amenable to single-molecule imaging.
Collapse
Affiliation(s)
- Kai Kikuchi
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Melbourne, VIC 305, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Liam D Adair
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.,School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jiarun Lin
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.,School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Elizabeth J New
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.,School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Amandeep Kaur
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Melbourne, VIC 305, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
74
|
Oberkersch RE, Lidonnici J, Santoro MM. How to Generate a Vascular-Labelled Transgenic Zebrafish Model to Study Tumor Angiogenesis and Extravasation. Methods Mol Biol 2023; 2572:191-202. [PMID: 36161418 DOI: 10.1007/978-1-0716-2703-7_15] [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: 06/16/2023]
Abstract
The use of transgenic animals carrying exogenous DNA integrated in their genome is a routine in modern-day laboratories. Nowadays, the zebrafish system represents the most useful tool for transgenesis studies mainly due to easy accessibility and manipulation of the eggs, which are produced in high numbers and over a relatively short generation time. The zebrafish transgenic technology is very straightforward when coupled with angiogenesis studies allowing easy in vivo observation of the vertebrate embryonic vasculature. Here, we describe the most common technique to generate vascular-labelled transgenic zebrafish embryos and their applications to study tumor angiogenesis and visualize tumor extravasation.
Collapse
Affiliation(s)
- Roxana E Oberkersch
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Jacopo Lidonnici
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
75
|
Moon H, Sultana T, Lee J, Huh J, Lee HD, Choi MS. Biomimetic lipid-fluorescein probe for cellular bioimaging. Front Chem 2023; 11:1151526. [PMID: 37153532 PMCID: PMC10160471 DOI: 10.3389/fchem.2023.1151526] [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: 01/27/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Fluorescence probe is one of the most powerful tools for cellular imaging. Here, three phospholipid-mimicking fluorescent probes (FP1-FP3) comprising fluorescein and two lipophilic groups of saturated and/or unsaturated C18 fatty acids were synthesized, and their optical properties were investigated. Like in biological phospholipids, the fluorescein group acts as a hydrophilic polar headgroup and the lipid groups act as hydrophobic non-polar tail groups. Laser confocal microscope images illustrated that FP3, which contains both saturated and unsaturated lipid tails, showed great uptake into the canine adipose-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Hyungkyu Moon
- Department of Materials Chemistry and Engineering, Konkuk University, Seoul, Republic of Korea
| | - Tania Sultana
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - JeongIk Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- *Correspondence: Myung-Seok Choi, ; JeongIk Lee,
| | - Jungrim Huh
- Social Eco-Tech Research Institute, Konkuk University, Seoul, Republic of Korea
| | - Hae Dong Lee
- Department of Materials Chemistry and Engineering, Konkuk University, Seoul, Republic of Korea
| | - Myung-Seok Choi
- Department of Materials Chemistry and Engineering, Konkuk University, Seoul, Republic of Korea
- *Correspondence: Myung-Seok Choi, ; JeongIk Lee,
| |
Collapse
|
76
|
Zhou C, Chia GWN, Yong KT. Membrane-intercalating conjugated oligoelectrolytes. Chem Soc Rev 2022; 51:9917-9932. [PMID: 36448452 DOI: 10.1039/d2cs00014h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
By acting as effective biomimetics of the lipid bilayers, membrane-intercalating conjugated oligoelectrolytes (MICOEs) can spontaneously insert themselves into both synthetic lipid bilayers and biological membranes. The modular and intentional molecular design of MICOEs enable a range of applications, such as bioproduction, biocatalysis, biosensing, and therapeutics. This tutorial review provides a structural evolution of MICOEs, which originated from the broader class of conjugated molecules, and analyses the drivers behind this evolutionary process. Various representative applications of MICOEs, accompanied by insights into their molecular design principles, will be reviewed separately. Perspectives on the current challenges and opportunities in research on MICOEs will be discussed at the end of the review to highlight their potential as unconventional and value-added materials for biological systems.
Collapse
Affiliation(s)
- Cheng Zhou
- Institute of Polymer Optoelectronic Materials and Devices, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, P. R. China. .,Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Geraldine W N Chia
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney 2006, New South Wales, Australia.
| |
Collapse
|
77
|
Mansuri S, Mukherjee T, Kanvah S. Fluorescent sterol probes for intracellular transport, imaging, and therapeutics. Curr Opin Chem Biol 2022; 71:102222. [PMID: 36219959 DOI: 10.1016/j.cbpa.2022.102222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 01/27/2023]
Abstract
Sterols play a significant role in many physiological processes affecting membrane organization, transport, permeability, and signal transduction. The development of fluorescent sterol analogs that have immediate functional relevance to the natural biomolecules is one approach to understanding the sterol-driven physiological processes. Visualizing cellular compartments with tailor-made fluorescent molecules through specific labeling methods enables organelle targeting and reveals dynamic information. In this review, we focus on the recent literature published between 2020 and 2022, with particular emphasis on extrinsic fluorophores and their investigations of sterol-driven biological processes involving sterol transport, biomolecular interactions, and biological imaging.
Collapse
Affiliation(s)
- Shabnam Mansuri
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382055, India
| | - Tarushyam Mukherjee
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382055, India
| | - Sriram Kanvah
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382055, India.
| |
Collapse
|
78
|
Driedonks T, Jiang L, Carlson B, Han Z, Liu G, Queen SE, Shirk EN, Gololobova O, Liao Z, Nyberg LH, Lima G, Paniushkina L, Garcia‐Contreras M, Schonvisky K, Castell N, Stover M, Guerrero‐Martin S, Richardson R, Smith B, Mahairaki V, Lai CP, Izzi JM, Hutchinson EK, Pate KAM, Witwer KW. Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e59. [PMID: 36591537 PMCID: PMC9799283 DOI: 10.1002/jex2.59] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/23/2022] [Accepted: 08/13/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) have potential in disease treatment since they can be loaded with therapeutic molecules and engineered for retention by specific tissues. However, questions remain on optimal dosing, administration, and pharmacokinetics. Previous studies have addressed biodistribution and pharmacokinetics in rodents, but little evidence is available for larger animals. Here, we investigated the pharmacokinetics and biodistribution of Expi293F-derived EVs labelled with a highly sensitive nanoluciferase reporter (palmGRET) in a non-human primate model (Macaca nemestrina), comparing intravenous (IV) and intranasal (IN) administration over a 125-fold dose range. We report that EVs administered IV had longer circulation times in plasma than previously reported in mice and were detectable in cerebrospinal fluid (CSF) after 30-60 minutes. EV association with PBMCs, especially B-cells, was observed as early as one minute post-administration. EVs were detected in liver and spleen within one hour of IV administration. However, IN delivery was minimal, suggesting that pretreatment approaches may be needed in large animals. Furthermore, EV circulation times strongly decreased after repeated IV administration, possibly due to immune responses and with clear implications for xenogeneic EV-based therapeutics. We hope that our findings from this baseline study in macaques will help to inform future research and therapeutic development of EVs.
Collapse
Affiliation(s)
- Tom Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zheng Han
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Guanshu Liu
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Erin N. Shirk
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zhaohao Liao
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lyle H. Nyberg
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Liliia Paniushkina
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Marta Garcia‐Contreras
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kayla Schonvisky
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mitchel Stover
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Selena Guerrero‐Martin
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Vasiliki Mahairaki
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Charles P. Lai
- Institute of Atomic and Molecular SciencesAcademia SinicaTaipeiTaiwan
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Eric K. Hutchinson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kelly A. M. Pate
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
79
|
Collot M, Pfister S, Klymchenko AS. Advanced functional fluorescent probes for cell plasma membranes. Curr Opin Chem Biol 2022; 69:102161. [DOI: 10.1016/j.cbpa.2022.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
|
80
|
Arifin DR, Witwer KW, Bulte JWM. Non-Invasive imaging of extracellular vesicles: Quo vaditis in vivo? J Extracell Vesicles 2022; 11:e12241. [PMID: 35844061 PMCID: PMC9289215 DOI: 10.1002/jev2.12241] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 01/11/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayer delimited vesicles released by nearly all cell types that serve as mediators of intercellular signalling. Recent evidence has shown that EVs play a key role in many normal as well as pathological cellular processes. EVs can be exploited as disease biomarkers and also as targeted, cell-free therapeutic delivery and signalling vehicles for use in regenerative medicine and other clinical settings. Despite this potential, much remains unknown about the in vivo biodistribution and pharmacokinetic profiles of EVs after administration into living subjects. The ability to non-invasively image exogeneous EVs, especially in larger animals, will allow a better understanding of their in vivo homing and retention patterns, blood and tissue half-life, and excretion pathways, all of which are needed to advance clinical diagnostic and/or therapeutic applications of EVs. We present the current state-of-the-art methods for labeling EVs with various diagnostic contrast agents and tracers and the respective imaging modalities that can be used for their in vivo visualization: magnetic resonance imaging (MRI), X-ray computed tomography (CT) imaging, magnetic particle imaging (MPI), single-photon emission computed tomography (SPECT), positron emission tomography (PET), and optical imaging (fluorescence and bioluminescence imaging). We review here the strengths and weaknesses of each of these EV imaging approaches, with special emphasis on clinical translation.
Collapse
Affiliation(s)
- Dian R. Arifin
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Neurologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jeff W. M. Bulte
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Oncologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical & Biomolecular Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
81
|
Salomon C, Das S, Erdbrügger U, Kalluri R, Kiang Lim S, Olefsky JM, Rice GE, Sahoo S, Andy Tao W, Vader P, Wang Q, Weaver AM. Extracellular Vesicles and Their Emerging Roles as Cellular Messengers in Endocrinology: An Endocrine Society Scientific Statement. Endocr Rev 2022; 43:441-468. [PMID: 35552682 PMCID: PMC10686249 DOI: 10.1210/endrev/bnac009] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 12/15/2022]
Abstract
During the last decade, there has been great interest in elucidating the biological role of extracellular vesicles (EVs), particularly, their hormone-like role in cell-to-cell communication. The field of endocrinology is uniquely placed to provide insight into the functions of EVs, which are secreted from all cells into biological fluids and carry endocrine signals to engage in paracellular and distal interactions. EVs are a heterogeneous population of membrane-bound vesicles of varying size, content, and bioactivity. EVs are specifically packaged with signaling molecules, including lipids, proteins, and nucleic acids, and are released via exocytosis into biofluid compartments. EVs regulate the activity of both proximal and distal target cells, including translational activity, metabolism, growth, and development. As such, EVs signaling represents an integral pathway mediating intercellular communication. Moreover, as the content of EVs is cell-type specific, it is a "fingerprint" of the releasing cell and its metabolic status. Recently, changes in the profile of EV and bioactivity have been described in several endocrine-related conditions including diabetes, obesity, cardiovascular diseases, and cancer. The goal of this statement is to highlight relevant aspects of EV research and their potential role in the field of endocrinology.
Collapse
Affiliation(s)
- Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Saumya Das
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Uta Erdbrügger
- Department of Medicine, Nephrology Division, University of Virginia, Charlottesville, VA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Jerrold M Olefsky
- Department of Medicine, University of California-San Diego, La Jolla, CA, USA
| | | | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Pieter Vader
- CDL Research, Division LAB, UMC Utrecht, Utrecht, the Netherlands Faculty of Medicine, Utrecht University, Utrecht, the Netherlands; Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, The Netherlands
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
82
|
Iyer RR, Liu YZ, Renteria CA, Tibble BE, Choi H, Žurauskas M, Boppart SA. Ultra-parallel label-free optophysiology of neural activity. iScience 2022; 25:104307. [PMID: 35602935 PMCID: PMC9114528 DOI: 10.1016/j.isci.2022.104307] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/18/2022] [Accepted: 04/22/2022] [Indexed: 01/21/2023] Open
Abstract
The electrical activity of neurons has a spatiotemporal footprint that spans three orders of magnitude. Traditional electrophysiology lacks the spatial throughput to image the activity of an entire neural network; besides, labeled optical imaging using voltage-sensitive dyes and tracking Ca2+ ion dynamics lack the versatility and speed to capture fast-spiking activity, respectively. We present a label-free optical imaging technique to image the changes to the optical path length and the local birefringence caused by neural activity, at 4,000 Hz, across a 200 × 200 μm2 region, and with micron-scale spatial resolution and 300-pm displacement sensitivity using Superfast Polarization-sensitive Off-axis Full-field Optical Coherence Microscopy (SPoOF OCM). The undulations in the optical responses from mammalian neuronal activity were matched with field-potential electrophysiology measurements and validated with channel blockers. By directly tracking the widefield neural activity at millisecond timescales and micrometer resolution, SPoOF OCM provides a framework to progress from low-throughput electrophysiology to high-throughput ultra-parallel label-free optophysiology.
Collapse
Affiliation(s)
- Rishyashring R. Iyer
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yuan-Zhi Liu
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Carlos A. Renteria
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brian E. Tibble
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Honggu Choi
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Mantas Žurauskas
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Corresponding author
| |
Collapse
|
83
|
Tkach M, Thalmensi J, Timperi E, Gueguen P, Névo N, Grisard E, Sirven P, Cocozza F, Gouronnec A, Martin-Jaular L, Jouve M, Delisle F, Manel N, Rookhuizen DC, Guerin CL, Soumelis V, Romano E, Segura E, Théry C. Extracellular vesicles from triple negative breast cancer promote pro-inflammatory macrophages associated with better clinical outcome. Proc Natl Acad Sci U S A 2022; 119:e2107394119. [PMID: 35439048 PMCID: PMC9169908 DOI: 10.1073/pnas.2107394119] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Tumor associated macrophages (TAMs), which differentiate from circulating monocytes, are pervasive across human cancers and comprise heterogeneous populations. The contribution of tumor-derived signals to TAM heterogeneity is not well understood. In particular, tumors release both soluble factors and extracellular vesicles (EVs), whose respective impact on TAM precursors may be different. Here, we show that triple negative breast cancer cells (TNBCs) release EVs and soluble molecules promoting monocyte differentiation toward distinct macrophage fates. EVs specifically promoted proinflammatory macrophages bearing an interferon response signature. The combination in TNBC EVs of surface CSF-1 promoting survival and cargoes promoting cGAS/STING or other activation pathways led to differentiation of this particular macrophage subset. Notably, macrophages expressing the EV-induced signature were found among patients’ TAMs. Furthermore, higher expression of this signature was associated with T cell infiltration and extended patient survival. Together, this data indicates that TNBC-released CSF-1-bearing EVs promote a tumor immune microenvironment associated with a better prognosis in TNBC patients.
Collapse
Affiliation(s)
- Mercedes Tkach
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Jessie Thalmensi
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Eleonora Timperi
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Paul Gueguen
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Nathalie Névo
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Eleonora Grisard
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Philemon Sirven
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Federico Cocozza
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Alizée Gouronnec
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | | | - Mabel Jouve
- CNRS UMR3215, Institut Curie, PSL Research University, 75005, Paris, France
| | - Fabien Delisle
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Nicolas Manel
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | | | - Coralie L. Guerin
- Cytometry Platform, CurieCoreTech, Institut Curie, Paris, F-75005 France
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, Paris, F-75006 France
| | - Vassili Soumelis
- Université de Paris, Inserm, U976 HIPI Unit, F-75006, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, F-75010, Paris, France
| | - Emanuela Romano
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Elodie Segura
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - Clotilde Théry
- INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| |
Collapse
|
84
|
Michelis S, Danglot L, Vauchelles R, Klymchenko AS, Collot M. Imaging and Measuring Vesicular Acidification with a Plasma Membrane-Targeted Ratiometric pH Probe. Anal Chem 2022; 94:5996-6003. [PMID: 35377610 DOI: 10.1021/acs.analchem.2c00574] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tracking the pH variation of intracellular vesicles throughout the endocytosis pathway is of prior importance to better assess the cell trafficking and metabolism of cells. Small molecular fluorescent pH probes are valuable tools in bioimaging but are generally not targeted to intracellular vesicles or are directly targeted to acidic lysosomes, thus not allowing the dynamic observation of the vesicular acidification. Herein, we designed Mem-pH, a fluorogenic ratiometric pH probe based on chromenoquinoline with appealing photophysical properties, which targets the plasma membrane (PM) of cells and further accumulates in the intracellular vesicles by endocytosis. The exposition of Mem-pH toward the vesicle's lumen allowed to monitor the acidification of the vesicles throughout the endocytic pathway and enabled the measurement of their pH via ratiometric imaging.
Collapse
Affiliation(s)
- Sophie Michelis
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Lydia Danglot
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, 75014 Paris, France
| | - Romain Vauchelles
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Mayeul Collot
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| |
Collapse
|
85
|
A near-infrared plasma membrane-specific AIE probe for fluorescence lifetime imaging of phagocytosis. Sci China Chem 2022. [DOI: 10.1007/s11426-021-1199-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AbstractPhagocytosis is a biological process that plays a key role in host defense and tissue homeostasis. Efficient approaches for realtime imaging of phagocytosis are highly desired but limited. Herein, an AIE-active near-infrared fluorescent probe, named TBTCP, was developed for fluorescence lifetime imaging of phagocytosis. TBTCP could selectively label the cell plasma membrane with fast staining, wash-free process, high signal-to-background ratio, and excellent photostability. Cellular membrane statuses under different osmolarities as well as macrophage phagocytosis of bacteria or large silica particles in early stages could be reported by the fluorescence lifetime changes of TBTCP. Compared with current fluorescence imaging methods, which target the bioenvironmental changes in the late phagocytosis stage, this approach detects the changes in the cell membrane, thus giving a faster response to phagocytosis. This article provides a functional tool to report the phagocytic dynamics of macrophages which may greatly contribute to the studies of phagocytic function-related diseases.
Collapse
|
86
|
Zhang X, Wang Z, Chu H, Xiong Z, Li Y, Chen Y, Zhu Q, Feng H, Zhu E, Zhou J, Huang P, Qian Z. Antipermeability Strategy to Achieve Extremely High Specificity and Ultralong Imaging of Diverse Cell Membranes Based on Restriction-Induced Emission of AIEgens. Anal Chem 2022; 94:4048-4058. [PMID: 35191676 DOI: 10.1021/acs.analchem.1c05345] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Long-term in situ cell membrane-targeted bioimaging is of great significance for studying specific biological processes and functions, but currently developed membrane probes are rarely simultaneously used to image the plasma membrane of animal and plant cells, and these probes lack sufficiently high long-term targeting ability. Herein, we proposed an antipermeability strategy to achieve highly specific and long-term imaging of plasma membranes of both human and plant cells using the steric hindrance effect and restriction-induced emission of AIE-active probes based on an updated membrane model. A certain degree of rigidity of plasma membrane containing a large ratio of rigid cholesterol molecules in the updated membrane model provides a promising opportunity to design antipermeable probes by introducing a rigid steric hindrance group in the probe. The designed antipermeable probes can anchor inside plasma membrane for a long term relying on the combination of the steric hindrance effect and the electrostatic and hydrophobic interactions between the probe and the membrane, as well as light up the membrane via the restriction-induced emission mechanism. The excellent performance in imaging completeness and specificity for both human cells and plant cells clearly shows that these designed probes possess outstanding antipermeability to achieve long-term specific imaging of membrane. These probes also show some advanced features such as ultrafast staining, wash-free merit, favorable biocompatibility, good photostability, and effective resistance to viscosity and pH alteration. This work also provides a valuable design principle for membrane probes of plant cells that the designed probes require a suitable molecular size favoring the penetration of small pores of cell walls.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Zhenni Wang
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Hao Chu
- College of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Zuping Xiong
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Yanjiang Li
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Yi Chen
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Qiaozhi Zhu
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Hui Feng
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Engao Zhu
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Jin Zhou
- College of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Peng Huang
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Zhaosheng Qian
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua 321004, People's Republic of China.,Key Laboratory of the Ministry for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| |
Collapse
|
87
|
Feng S, Liu Y, Li Q, Gui Z, Feng G. Two Water-Soluble and Wash-Free Fluorogenic Probes for Specific Lighting Up Cancer Cell Membranes and Tumors. Anal Chem 2022; 94:1601-1607. [PMID: 35015515 DOI: 10.1021/acs.analchem.1c03685] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The construction of microenvironment-sensitive probes with good cell membrane-targetability can reveal the fundamental properties of cell membranes. Herein, two polarity-sensitive probes, termed MEMs were reported for the first time to specifically light up cancer cell membranes. Both probes were designed with tetrahydroquinoxaline coumarin amide as the fluorophore, and quaternary ammonium groups were appended to increase water solubility and target cell membranes. In vitro studies showed that the fluorescence of both probes displayed strong polarity dependence and had a wide linear range to polarity (Δf). MEMs also displayed excellent cell membrane targeting ability and could long-term light up cell membranes with red fluorescence and a wash-free process. More excitingly, MEMs could specifically light up cancer cell membranes, revealing that cancer cells might have lower cell membrane polarity than normal cells. In vivo studies showed that MEMs could also effectively distinguish tumors from normal tissues. Overall, this work has not only developed two polarity-sensitive probes with good cell membrane targetability, but also provided new insights and methods for an in-depth understanding of cancer cells and cancer diagnosis.
Collapse
Affiliation(s)
- Shumin Feng
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| | - Yijia Liu
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| | - Qianhua Li
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| | - Zhisheng Gui
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| | - Guoqiang Feng
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| |
Collapse
|
88
|
Melling GE, Conlon R, Pantazi P, Dellar ER, Samuel P, Baena-Lopez LA, Simpson JC, Carter DRF. Confocal microscopy analysis reveals that only a small proportion of extracellular vesicles are successfully labelled with commonly utilised staining methods. Sci Rep 2022; 12:262. [PMID: 34997141 PMCID: PMC8741769 DOI: 10.1038/s41598-021-04225-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022] Open
Abstract
Assessing genuine extracellular vesicle (EV) uptake is crucial for understanding the functional roles of EVs. This study measured the bona fide labelling of EVs utilising two commonly used fluorescent dyes, PKH26 and C5-maleimide-Alexa633. MCF7 EVs tagged with mEmerald-CD81 were isolated from conditioned media by size exclusion chromatography (SEC) and characterised using Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM), MACsPlex immunocapture assay and immunoblots. These fluorescently tagged EVs were subsequently stained with C5-maleimide-Alexa633 or PKH26, according to published protocols. Colocalisation of dual-labelled EVs was assessed by confocal microscopy and quantified using the Rank-Weighted Colocalisation (RWC) algorithm. We observed strikingly poor colocalisation between mEmerald-CD81-tagged EVs and C5-Maleimide-Alexa633 (5.4% ± 1.8) or PKH26 (4.6% ± 1.6), that remained low even when serum was removed from preparations. Our data confirms previous work showing that some dyes form contaminating aggregates. Furthermore, uptake studies showed that maleimide and mEmerald-CD81-tagged EVs can be often located into non-overlapping subcellular locations. By using common methods to isolate and stain EVs we observed that most EVs remained unstained and most dye signal does not appear to be EV associated. Our work shows that there is an urgent need for optimisation and standardisation in how EV researchers use these tools to assess genuine EV signals.
Collapse
Affiliation(s)
- Genevieve E Melling
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
- Institute of Clinical Sciences, School of Biomedical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ross Conlon
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Science Centre West, Belfield, Dublin 4, Ireland
| | - Paschalia Pantazi
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, UK
| | - Elizabeth R Dellar
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Priya Samuel
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
| | - Luis Alberto Baena-Lopez
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Jeremy C Simpson
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Science Centre West, Belfield, Dublin 4, Ireland.
| | - David R F Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK.
- Evox Therapeutics Ltd, Oxford Science Park, Medawar Centre, Robert Robinson Avenue, Oxford, OX4 4HG, UK.
| |
Collapse
|
89
|
Ren Y, Ge K, Sun D, Hong Z, Jia C, Hu H, Shao F, Yao B. Rapid enrichment and sensitive detection of extracellular vesicles through measuring the phospholipids and transmembrane protein in a microfluidic chip. Biosens Bioelectron 2021; 199:113870. [PMID: 34915212 DOI: 10.1016/j.bios.2021.113870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 11/02/2022]
Abstract
Extracellular vesicles (EVs) have attracted tremendous attention in recent years and quantification of EVs is a key issue in the evaluation of vesicle-based diagnostics and therapeutic development, but it's quite challenging to determine whether higher protein expression signals are due to larger vesicle amount or higher protein content within each vesicle. To solve this problem, herein, we proposed a strategy based on staining phospholipid bilayers of EVs with lipophilic dyes to evaluate their lipid amount, which was subsequently normalized as an internal standard for studying the expression of transmembrane protein (i.e., CD63) on EVs in different samples. In addition, a microfluidic platform based on electrophoresis technology was invented to effectively enrich and detect EVs. Small fluorescent labeling molecules (i.e., uncombined aptamers) were on-chip removed from EVs without pre-separation via ultracentrifugation or ultrafiltration which were indispensable in nanoparticle tracking analysis (NTA) and flow cytometry techniques and the performance of this assay is comparable to NTA. Finally, it was found obvious difference in the expression of CD63 on EVs before and after normalization based on lipid amount in plasma samples. This method is expected to provide more accurate information when comparing the expression levels of EVs biomarkers in different samples.
Collapse
Affiliation(s)
- Yongan Ren
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Ke Ge
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, The Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, China
| | - Danyang Sun
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Changku Jia
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, The Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, China
| | - Huan Hu
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, China
| | - Fangwei Shao
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou, China.
| |
Collapse
|
90
|
Dadina N, Tyson J, Zheng S, Lesiak L, Schepartz A. Imaging organelle membranes in live cells at the nanoscale with lipid-based fluorescent probes. Curr Opin Chem Biol 2021; 65:154-162. [PMID: 34715587 PMCID: PMC9904808 DOI: 10.1016/j.cbpa.2021.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022]
Abstract
Understanding how organelles interact, exchange materials, assemble, disassemble, and evolve as a function of space, time, and environment is an exciting area at the very forefront of chemical and cell biology. Here, we bring attention to recent progress in the design and application of lipid-based tools to visualize and interrogate organelles in live cells, especially at super resolution. We highlight strategies that rely on modification of natural lipids or lipid-like small molecules ex cellula, where organelle specificity is provided by the structure of the chemically modified lipid, or in cellula using cellular machinery, where an enzyme labels the lipid in situ. We also describe recent improvements to the chemistry upon which lipid probes rely, many of which have already begun to broaden the scope of biological questions that can be addressed by imaging organelle membranes at the nanoscale.
Collapse
Affiliation(s)
- N. Dadina
- Department of Chemistry, University of California, Berkeley 94720, USA
| | - J. Tyson
- Department of Chemistry, University of California, Berkeley 94720, USA
| | - S. Zheng
- Department of Chemistry, University of California, Berkeley 94720, USA
| | - L. Lesiak
- Department of Chemistry, University of California, Berkeley 94720, USA
| | - A. Schepartz
- Department of Chemistry, University of California, Berkeley 94720, USA,Department of Molecular & Cell Biology, University of California, Berkeley 94720, USA,California Institute for Quantitative Biosciences, University of California, Berkeley 94720, USA
| |
Collapse
|
91
|
Show your true color: Mammalian cell surface staining for tracking cellular identity in multiplexing and beyond. Curr Opin Chem Biol 2021; 66:102102. [PMID: 34861482 DOI: 10.1016/j.cbpa.2021.102102] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 12/23/2022]
Abstract
Fluorescence microscopy revolutionized cell biology and changed requirements for dyes towards higher brightness, novel capacities, and specific targets. With the need for multiplexing assays in high-throughput methodologies, surface staining gained particular interest because it allows rapid application of exogenous stains to track cellular identity in mixed populations. Indeed, the last decade has enriched the toolbox of general lipid stains, fluorescent lipid analogues, sugar-binding lectins, and protein-specific antibodies enabling the first rationally designed plasma membrane-specific dyes. Still, multiple challenges exist, and the unique properties of each dye must be considered when selecting a staining approach for a specific application. Recent advances are also promising that future dyes will provide ultimate brightness and photostability in diverse colors and reduced sizes for high-resolution imaging.
Collapse
|
92
|
A near-infrared AIE fluorescent probe for myelin imaging: From sciatic nerve to the optically cleared brain tissue in 3D. Proc Natl Acad Sci U S A 2021; 118:2106143118. [PMID: 34740969 PMCID: PMC8609329 DOI: 10.1073/pnas.2106143118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 12/25/2022] Open
Abstract
The high spatial resolution of three-dimensional (3D) fluorescence imaging of myelinated fibers will greatly facilitate the understanding of 3D neural networks and the pathophysiology of demyelinating diseases. However, existing myelin probes are far from satisfactory because of their low–signal-to-background ratio and poor tissue permeability. We herein developed a near-infrared aggregation-induced emission-active probe, PM-ML, for high-performance myelin imaging. PM-ML could specifically image myelinated fibers in teased sciatic nerves and mouse brain tissues with high contrast, good photostability, and deep penetration depth. PM-ML staining is compatible with several tissue-clearing methods. Its application in assessing myelination for neuropathological studies was also demonstrated using a multiple sclerosis mouse model. Myelin, the structure that surrounds and insulates neuronal axons, is an important component of the central nervous system. The visualization of the myelinated fibers in brain tissues can largely facilitate the diagnosis of myelin-related diseases and understand how the brain functions. However, the most widely used fluorescent probes for myelin visualization, such as Vybrant DiD and FluoroMyelin, have strong background staining, low-staining contrast, and low brightness. These drawbacks may originate from their self-quenching properties and greatly limit their applications in three-dimensional (3D) imaging and myelin tracing. Chemical probes for the fluorescence imaging of myelin in 3D, especially in optically cleared tissue, are highly desirable but rarely reported. We herein developed a near-infrared aggregation-induced emission (AIE)-active probe, PM-ML, for high-performance myelin imaging. PM-ML is plasma membrane targeting with good photostability. It could specifically label myelinated fibers in teased sciatic nerves and mouse brain tissues with a high–signal-to-background ratio. PM-ML could be used for 3D visualization of myelin sheaths, myelinated fibers, and fascicles with high-penetration depth. The staining is compatible with different brain tissue–clearing methods, such as ClearT and ClearT2. The utility of PM-ML staining in demyelinating disease studies was demonstrated using the mouse model of multiple sclerosis. Together, this work provides an important tool for high-quality myelin visualization across scales, which may greatly contribute to the study of myelin-related diseases.
Collapse
|
93
|
Optimization of Advanced Live-Cell Imaging through Red/Near-Infrared Dye Labeling and Fluorescence Lifetime-Based Strategies. Int J Mol Sci 2021; 22:ijms222011092. [PMID: 34681761 PMCID: PMC8537913 DOI: 10.3390/ijms222011092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/30/2022] Open
Abstract
Fluorescence microscopy is essential for a detailed understanding of cellular processes; however, live-cell preservation during imaging is a matter of debate. In this study, we proposed a guide to optimize advanced light microscopy approaches by reducing light exposure through fluorescence lifetime (τ) exploitation of red/near-infrared dyes. Firstly, we characterized key instrumental elements which revealed that red/near-infrared laser lines with an 86x (Numerical Aperture (NA) = 1.2, water immersion) objective allowed high transmission of fluorescence signals, low irradiance and super-resolution. As a combination of two technologies, i.e., vacuum tubes (e.g., photomultiplier) and semiconductor microelectronics (e.g., avalanche photodiode), type S, X and R of hybrid detectors (HyD-S, HyD-X and HyD-R) were particularly adapted for red/near-infrared photon counting and τ separation. Secondly, we tested and compared lifetime-based imaging including coarse τ separation for confocal microscopy, fitting and phasor plot analysis for fluorescence lifetime microscopy (FLIM), and lifetimes weighting for enhanced stimulated emission depletion (STED) nanoscopy, in light of red/near-infrared multiplexing. Mainly, we showed that the choice of appropriate imaging approach may depend on fluorochrome number, together with their spectral/lifetime characteristics and STED compatibility. Photon-counting mode and sensitivity of HyDs together with phasor plot analysis of fluorescence lifetimes enabled the flexible and fast imaging of multi-labeled living H28 cells. Therefore, a combination of red/near-infrared dyes labeling with lifetime-based strategies offers new perspectives for live-cell imaging by enhancing sample preservation through acquisition time and light exposure reduction.
Collapse
|
94
|
Abstract
Fluorescence imaging techniques play a pivotal role in our understanding of the nervous system. The emergence of various super-resolution microscopy methods and specialized fluorescent probes enables direct insight into neuronal structure and protein arrangements in cellular subcompartments with so far unmatched resolution. Super-resolving visualization techniques in neurons unveil a novel understanding of cytoskeletal composition, distribution, motility, and signaling of membrane proteins, subsynaptic structure and function, and neuron-glia interaction. Well-defined molecular targets in autoimmune and neurodegenerative disease models provide excellent starting points for in-depth investigation of disease pathophysiology using novel and innovative imaging methodology. Application of super-resolution microscopy in human brain samples and for testing clinical biomarkers is still in its infancy but opens new opportunities for translational research in neurology and neuroscience. In this review, we describe how super-resolving microscopy has improved our understanding of neuronal and brain function and dysfunction in the last two decades.
Collapse
Affiliation(s)
- Christian Werner
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
95
|
Mukherjee T, Kanvah S, Klymchenko AS, Collot M. Probing Variations of Reduction Activity at the Plasma Membrane Using a Targeted Ratiometric FRET Probe. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40315-40324. [PMID: 34424677 DOI: 10.1021/acsami.1c11069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasma membrane (PM) is the turntable of various reactions that regulate essential functionalities of cells. Among these reactions, the thiol disulfide exchange (TDE) reaction plays an important role in cellular processes. We herein designed a selective probe, called membrane reduction probe (MRP), that is able to report TDE activity at the PM. MRP is based on a green emitting BODIPY PM probe connected to rhodamine through a disulfide bond. MRP is fluorogenic as it is turned off in aqueous media due to aggregation-caused quenching, and once inserted in the PM, it displays a bright red signal due to an efficient fluorescence energy resonance transfer (FRET) between the BODIPY donor and the rhodamine acceptor. In the PM model, the MRP can undergo TDE reaction with external reductive agents as well as with thiolated lipids embedded in the bilayer. Upon TDE reaction, the FRET is turned off and a bright green signal appears allowing a ratiometric readout of this reaction. In cells, the MRP quickly labeled the PM and was able to probe variations of TDE activity using ratiometric imaging. With this tool in hand, we were able to monitor variations of TDE activity at the PM under stress conditions, and we showed that cancer cell lines presented a reduced TDE activity at the PM compared to noncancer cells.
Collapse
Affiliation(s)
- Tarushyam Mukherjee
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Sriram Kanvah
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Mayeul Collot
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| |
Collapse
|
96
|
Verweij FJ, Balaj L, Boulanger CM, Carter DRF, Compeer EB, D'Angelo G, El Andaloussi S, Goetz JG, Gross JC, Hyenne V, Krämer-Albers EM, Lai CP, Loyer X, Marki A, Momma S, Nolte-'t Hoen ENM, Pegtel DM, Peinado H, Raposo G, Rilla K, Tahara H, Théry C, van Royen ME, Vandenbroucke RE, Wehman AM, Witwer K, Wu Z, Wubbolts R, van Niel G. The power of imaging to understand extracellular vesicle biology in vivo. Nat Methods 2021; 18:1013-1026. [PMID: 34446922 PMCID: PMC8796660 DOI: 10.1038/s41592-021-01206-3] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/20/2021] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized lipid bilayer vesicles released by virtually every cell type. EVs have diverse biological activities, ranging from roles in development and homeostasis to cancer progression, which has spurred the development of EVs as disease biomarkers and drug nanovehicles. Owing to the small size of EVs, however, most studies have relied on isolation and biochemical analysis of bulk EVs separated from biofluids. Although informative, these approaches do not capture the dynamics of EV release, biodistribution, and other contributions to pathophysiology. Recent advances in live and high-resolution microscopy techniques, combined with innovative EV labeling strategies and reporter systems, provide new tools to study EVs in vivo in their physiological environment and at the single-vesicle level. Here we critically review the latest advances and challenges in EV imaging, and identify urgent, outstanding questions in our quest to unravel EV biology and therapeutic applications.
Collapse
Affiliation(s)
- Frederik J Verweij
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - David R F Carter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
- Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Ewoud B Compeer
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Gisela D'Angelo
- Institut Curie, PSL Research University, CNRS, UMR144 Cell Biology and Cancer, Paris, France
| | - Samir El Andaloussi
- Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics Lab, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Equipe Labellisée Ligue contre le Cancer, Strasbourg, France
| | | | - Vincent Hyenne
- INSERM UMR_S1109, Tumor Biomechanics Lab, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Equipe Labellisée Ligue contre le Cancer, Strasbourg, France
- CNRS SNC5055, Strasbourg, France
| | - Eva-Maria Krämer-Albers
- Johannes Gutenberg-Universität Mainz, Institute of Developmental Biology and Neurobiology, Mainz, Germany
| | - Charles P Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Xavier Loyer
- Université de Paris, PARCC, INSERM, Paris, France
| | - Alex Marki
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Goethe-University, Frankfurt am Main, Germany
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Faculty of veterinary medicine, Utrecht University, Utrecht, the Netherlands
| | - D Michiel Pegtel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hector Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144 Cell Biology and Cancer, Paris, France
| | - Kirsi Rilla
- University of Eastern Finland, Institute of Biomedicine, Kuopio, Finland
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Immunity and Cancer, Paris, France
| | | | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ann M Wehman
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology and Neurology and the Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Richard Wubbolts
- Department of Biomolecular Health Sciences, Faculty of veterinary medicine, Utrecht University, Utrecht, the Netherlands
| | - Guillaume van Niel
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| |
Collapse
|
97
|
Fam KT, Saladin L, Klymchenko AS, Collot M. Confronting molecular rotors and self-quenched dimers as fluorogenic BODIPY systems to probe biotin receptors in cancer cells. Chem Commun (Camb) 2021; 57:4807-4810. [PMID: 33982709 DOI: 10.1039/d1cc00108f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Probing receptors at the cell surface to monitor their expression level can be performed with fluorogenic dyes. Biotin receptors (BRs) are particularly interesting as they are overexpressed in cancer cells. Herein, to image BRs, we adapted and systematically compared two fluorogenic systems based on BODIPYs: a molecular rotor and a self-quenched dimer that light up in response to high viscosity and low polarity of the membrane, respectively. The fluorogenic dimer proved to be more efficient than the rotor and allowed BRs to be imaged in cancer cells, which can effectively be discriminated from non-cancer cells.
Collapse
Affiliation(s)
- Kyong T Fam
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, Illkirch-Graffenstaden 67401, France.
| | - Lazare Saladin
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, Illkirch-Graffenstaden 67401, France.
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, Illkirch-Graffenstaden 67401, France.
| | - Mayeul Collot
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, Illkirch-Graffenstaden 67401, France.
| |
Collapse
|
98
|
Broichhagen J, Kilian N. Chemical Biology Tools To Investigate Malaria Parasites. Chembiochem 2021; 22:2219-2236. [PMID: 33570245 PMCID: PMC8360121 DOI: 10.1002/cbic.202000882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Parasitic diseases like malaria tropica have been shaping human evolution and history since the beginning of mankind. After infection, the response of the human host ranges from asymptomatic to severe and may culminate in death. Therefore, proper examination of the parasite's biology is pivotal to deciphering unique molecular, biochemical and cell biological processes, which in turn ensure the identification of treatment strategies, such as potent drug targets and vaccine candidates. However, implementing molecular biology methods for genetic manipulation proves to be difficult for many parasite model organisms. The development of fast and straightforward applicable alternatives, for instance small-molecule probes from the field of chemical biology, is essential. In this review, we will recapitulate the highlights of previous molecular and chemical biology approaches that have already created insight and understanding of the malaria parasite Plasmodium falciparum. We discuss current developments from the field of chemical biology and explore how their application could advance research into this parasite in the future. We anticipate that the described approaches will help to close knowledge gaps in the biology of P. falciparum and we hope that researchers will be inspired to use these methods to gain knowledge - with the aim of ending this devastating disease.
Collapse
Affiliation(s)
- Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)Robert-Roessle-Strasse 1013125BerlinGermany
| | - Nicole Kilian
- Centre for Infectious DiseasesParasitologyHeidelberg University HospitalIm Neuenheimer Feld 32469120HeidelbergGermany
| |
Collapse
|
99
|
Ashoka AH, Klymchenko AS. Ultrabright Fluorescent Polymeric Nanofibers and Coatings Based on Ionic Dye Insulation with Bulky Counterions. ACS APPLIED MATERIALS & INTERFACES 2021; 13:28889-28898. [PMID: 34106696 DOI: 10.1021/acsami.1c06436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Preparation of bright fluorescent materials based on polymers is hampered by a fundamental problem of aggregation-caused quenching (ACQ) of encapsulated dyes. Here, ultrabright fluorescent polymeric nanofibers and coatings are prepared based on a concept of ionic dye insulation with bulky hydrophobic counterions that overcomes the ACQ problem. It is found that bulky hydrophobic counterion perfluorinated tetraphenylborate can boost >100-fold the fluorescence quantum yields of cationic dye octadecyl rhodamine B at high loading (30 wt %) in biocompatible poly(methyl methacrylate) (PMMA). The concept is applicable to both rhodamine and cyanine dyes, which results in bright fluorescent polymeric materials of four different colors spanning from blue to near-infrared. It allows for preparation of electrospun polymeric nanofibers with >50-fold higher dye loading by mass (30 wt %, >20-fold higher molarity for rhodamine dyes) while preserving good fluorescence quantum yields (31%), which implies drastic improvement in their fluorescence brightness. The counterion-based polymeric materials are also validated as coatings of model medical devices, such as stainless steel fiducials and 3D-printed stents of complex geometry. Spin-coated fluorescent polymeric films loaded with a dye paired with bulky counterions exhibit excellent biocompatibility and low toxicity. Moreover, counterion-modified materials show much better stability against dye leakage in the presence of living cells and a serum-containing medium, compared to materials based on the dye with a small inorganic anion. Overall, by pushing the barriers of ACQ, our counterion approach emerges as a powerful tool to develop ultrabright fluorescent polymeric materials ranging from nano- and macroscale.
Collapse
Affiliation(s)
- Anila Hoskere Ashoka
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, 67401 Illkirch, France
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, 67401 Illkirch, France
| |
Collapse
|
100
|
Sikora R, Bun P, Danglot L, Alqabandi M, Bassereau P, Niedergang F, Galli T, Zahraoui A. MICAL-L1 is required for cargo protein delivery to the cell surface. Biol Open 2021; 10:269021. [PMID: 34100897 PMCID: PMC8214422 DOI: 10.1242/bio.058008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/22/2021] [Indexed: 11/21/2022] Open
Abstract
Secreted proteins are transported along intracellular route from the endoplasmic reticulum through the Golgi before reaching the plasma membrane. Small GTPase Rab and their effectors play a key role in membrane trafficking. Using confocal microscopy, we showed that MICAL-L1 was associated with tubulo-vesicular structures and exhibited a significant colocalization with markers of the Golgi apparatus and recycling endosomes. Super resolution STORM microscopy suggested at the molecular level, a very close association of MICAL-L1 and microdomains in the Golgi cisternae. Using a synchronized secretion assay, we report that the shRNA-mediated depletion of MICAL-L1 impaired the delivery of a subset of cargo proteins to the cell surface. The process of membrane tubulation was monitored in vitro, and we observe that recombinant MICAL-L1-RBD domain may contribute to promote PACSINs-mediated membrane tubulation. Interestingly, two hydrophobic residues at the C-terminus of MICAL-L1 appeared to be important for phosphatidic acid binding, and for association with membrane tubules. Our results reveal a new role for MICAL-L1 in cargo delivery to the plasma membrane. Summary: MICAL-L1, an effector of Rab GTPases, exhibits a significant colocalization with markers of the Golgi apparatus and recycling endosomes. It is involved in cargo delivery to the plasma membrane.
Collapse
Affiliation(s)
- R Sikora
- Université de Paris, Inserm U1016-CNRS UMR 8104, Institut Cochin, Paris, France
| | - P Bun
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging facility, 75014 Paris, France
| | - L Danglot
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging facility, 75014 Paris, France
| | - M Alqabandi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS, UMR168, 75005, Paris, France
| | - P Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS, UMR168, 75005, Paris, France
| | - F Niedergang
- Université de Paris, Inserm U1016-CNRS UMR 8104, Institut Cochin, Paris, France
| | - T Galli
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France.,GHU PARIS psychiatrie & neurosciences, F-75014 Paris, France
| | - A Zahraoui
- Université de Paris, Inserm U1016-CNRS UMR 8104, Institut Cochin, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France
| |
Collapse
|