1
|
Elnegris HM, Abdelrahman AA, El-Roghy ES. The potential therapeutic effects of exosomes derived from bone marrow mesenchymal stem cells on ileum injury of a rat sepsis model (histological and immunohistochemical study). Ultrastruct Pathol 2024; 48:274-296. [PMID: 38946300 DOI: 10.1080/01913123.2024.2368011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Sepsis denotes a serious high mortality concern. The study was designed to evaluate the effect of mesenchymal stem cell exosomes (MSC-exosomes) on the evolution of the animal model of sepsis. In this study, 36 rats were distributed into three groups, (I) controls, (II) LPS-treated, and (III) LPS+MSC-EVs. Sepsis was simulated by administering E. coli-LPS to the laboratory animals. Group III was given MSC-exosomes four hours after the LPS injection. Forty-eight hours later rats were sacrificed. Ileum samples were excised, and processed for the histological assessment, immunohistochemical identification of CD44, and inducible nitric oxide synthase (iNOS). Ileum homogenate was used to estimate tumor necrosis factor α (TNF α) besides Cyclooxygenase-2 (COX 2). PCR was used for the detection of interleukin 1α (IL‑1α), and interleukin 17 (IL‑17). Statistical and morphometrical analysis was done. The LPS-treated group showed increased TNF-α, IL‑1α, IL‑17, and decreased COX 2. LPS administration led to cytoplasmic vacuolization of enterocytes, an increase in the vasculature, and cellular infiltrations invaded the lamina propria. There was a significant rise in goblet cells and the proportion of collagen fibers. Ultrastructurally, the enterocytes displayed nuclear irregularity, rough endoplasmic reticulum (rER) dilatation, and increased mitochondria number. Sepsis induces a significant increase in iNOS and a decrease in CD44 immune expressions. LPS+MSC-EVs group restored normal ileum structure and revealed a significant elevation in CD44 and a reduction in iNOS immunoreactions. LPS-sepsis induced an obvious ileum inflammatory deterioration ameliorated by MSC-exosomes, mostly through their antioxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Heba M Elnegris
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Department of Histology and Cell Biology, Faculty of Medicine, Badr University in Cairo, Cairo, Egypt
| | - Abeer A Abdelrahman
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman S El-Roghy
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shebin el Kom, Egypt
| |
Collapse
|
2
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [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: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
3
|
Liwski R, Greenshields A, Grace I, Rourke C, Cheng C, Quinn JG. The use of intracellular dyes to create a multiplexed flow cytometry-based red blood cell phenotyping assay. Vox Sang 2024; 119:752-757. [PMID: 38587165 DOI: 10.1111/vox.13630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/13/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND AND OBJECTIVES Flow cytometry can be used to phenotype red blood cell antigens, allowing for high-throughput testing while using low reagent volumes. This article utilizes intracellular dyes to pre-label red blood cells to further multiplex flow cytometry-based red blood cell antigen phenotyping. MATERIALS AND METHODS Red blood cells were pre-labelled using the intracellular dyes V450 and Oregon Green. These dyes are detected fluorescently via flow cytometry. Four combinations of intracellular staining were used to allow four patient or donor red blood cells to be analysed in a single test well. Antigen phenotyping was then performed via flow cytometry using a previously described method. RESULTS The intracellular dyes showed uniform staining when measured in mean fluorescence intensity and allowed the red blood cells to be clearly distinguished from one another. The presence or absence of red blood cell antigens was determined with 100% accuracy. CONCLUSION The use of intracellular dyes allowed a fourfold increase in the throughput of our previously described flow cytometry-based red blood cell antigen phenotyping method. The described method allows up to 48 patients to be simultaneously phenotyped using a single 96-well microplate. Furthermore, additional fluorescent dyes could potentially increase the throughput exponentially.
Collapse
Affiliation(s)
- Robert Liwski
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Anna Greenshields
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Ian Grace
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Craig Rourke
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Calvino Cheng
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Jason George Quinn
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pathology and Laboratory Medicine, Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| |
Collapse
|
4
|
Soh KT, Tario JD, Muirhead KA, Wallace PK. Probing cell proliferation: Considerations for dye selection. Methods Cell Biol 2024; 186:1-24. [PMID: 38705595 DOI: 10.1016/bs.mcb.2024.02.012] [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: 05/07/2024]
Abstract
Broadly speaking, cell tracking dyes are fluorescent compounds that bind stably to components on or within the cells so the fate of the labeled cells can be followed. Their staining should be bright and homogeneous without affecting cell function. For purposes of monitoring cell proliferation, each time a cell divides the intensity of cell tracking dye should diminish equally between daughter cells. These dyes can be grouped into two different classes. Protein reactive dyes label cells by reacting covalently but non-selectively with intracellular proteins. Carboxyfluorescein diacetate succinimidyl ester (CFSE) is the prototypic general protein label. Membrane intercalating dyes label cells by partitioning non-selectively and non-covalently within the plasma membrane. The PKH membrane dyes are examples of lipophilic compounds whose chemistry allows for their retention within biological membranes without affecting cellular growth, viability, or proliferation when used properly. Here we provide considerations based for labeling cell lines and peripheral blood mononuclear cells using both classes of dyes. Examples from optimization experiments are presented along with critical aspects of the staining procedures to help mitigate common risks. Of note, we present data where a logarithmically growing cell line is labeled with both a protein dye and a membrane tracking dye to compare dye loss rates over 6days. We found that dual stained cells paralleled dye loss of the corresponding single stained cells. The decrease in fluorescence intensity by protein reactive dyes, however, was more rapid than that with the membrane reactive dyes, indicating the presence of additional division-independent dye loss.
Collapse
Affiliation(s)
- Kah Teong Soh
- Agenus, Inc., Lexington, MA, United States; Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Joseph D Tario
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | | | - Paul K Wallace
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States; SciGro, Inc., Middleton, WI, United States.
| |
Collapse
|
5
|
Liwski R, Greenshields A, Grace I, Cheng C, Quinn JG. Development of multiplexed flow cytometry-based red blood cell antibody screen and identification assays. Vox Sang 2024; 119:344-352. [PMID: 38175141 DOI: 10.1111/vox.13584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/06/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND AND OBJECTIVES The purpose of this study was to develop a high-throughput method of performing red blood cell antibody screens and identification by utilizing flow cytometry and intracellular dyes to allow a multiplexed assay where three-cell screens can be performed in a single test well and 11-cell panels in three test wells. MATERIALS AND METHODS Reagent red blood cells were labelled using Violet Proliferation Dye 450 (V450) and Oregon Green fluorescent dyes, which bind intracellular proteins to allow up to four cells to be interrogated in a single test well. Sixteen 3-cell screen panels and ten 11-cell identification panels were tested using sera with known antibody specificity. Antibody binding was detected using secondary anti-immunoglobulin G and anti-immunoglobulin M fluorescently labelled antibodies. RESULTS Intracellular dyes allowed clear separation of the different screen and identification panel test cells. Three distinct populations of V450+, Oregon Green+ and negative for both stains were demonstrated in the screening panel and an additional double positive for V450 and Oregon Green was utilized to include a fourth cell in the identification panel testing to increase throughput. A total of 158 screen or identification panel RBC/serum combinations were tested against different known antibodies, and expected results were obtained with 100% concordance. CONCLUSION This study demonstrates the successful development of a high-throughput multiplexed flow cytometry-based red cell antibody screen and identification panel assays. This method could be implemented in clinical laboratories to complement existing antibody detection methods. The multiplexing enabled via intracellular staining could be utilized to further augment other flow cytometry-based transfusion assays.
Collapse
Affiliation(s)
- Robert Liwski
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Anna Greenshields
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Ian Grace
- Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Calvino Cheng
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| | - Jason George Quinn
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Central Zone, Halifax, Nova Scotia, Canada
| |
Collapse
|
6
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
7
|
Tario JD, Soh KT, Wallace PK, Muirhead KA. Monitoring Cell Proliferation by Dye Dilution: Considerations for Panel Design. Methods Mol Biol 2024; 2779:159-216. [PMID: 38526787 DOI: 10.1007/978-1-0716-3738-8_9] [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: 03/27/2024]
Abstract
High dimensional studies that include proliferation dyes face two inherent challenges in panel design. First, the more rounds of cell division to be monitored based on dye dilution, the greater the starting intensity of the labeled parent cells must be in order to distinguish highly divided daughter cells from background autofluorescence. Second, the greater their starting intensity, the more difficult it becomes to avoid spillover of proliferation dye signal into adjacent spectral channels, with resulting limitations on the use of other fluorochromes and ability to resolve dim signals of interest. In the third and fourth editions of this series, we described the similarities and differences between protein-reactive and membrane-intercalating dyes used for general cell tracking, provided detailed protocols for optimized labeling with each dye type, and summarized characteristics to be tested by the supplier and/or user when validating either dye type for use as a proliferation dye. In this fifth edition, we review: (a) Fundamental assumptions and critical controls for dye dilution proliferation assays; (b) Methods to evaluate the effect of labeling on cell growth rate and test the fidelity with which dye dilution reports cell division; and. (c) Factors that determine how many daughter generations can be accurately included in proliferation modeling. We also provide an expanded section on spectral characterization, using data collected for three protein-reactive dyes (CellTrace™ Violet, CellTrace™ CFSE, and CellTrace™ Far Red) and three membrane-intercalating dyes (PKH67, PKH26, and CellVue® Claret) on three different cytometers to illustrate typical decisions and trade-offs required during multicolor panel design. Lastly, we include methods and controls for assessing regulatory T cell potency, a functional assay that incorporates the "know your dye" and "know your cytometer" principles described herein.
Collapse
Affiliation(s)
- Joseph D Tario
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kah Teong Soh
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Agenus, Inc., Lexington, MA, USA
| | - Paul K Wallace
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- SciGro, Inc., Sedona, AZ, USA
| | | |
Collapse
|
8
|
Priyadarsini M, Kushwaha J, Pandey KP, Rani J, Dhoble AS. Application of flow cytometry for rapid, high-throughput, multiparametric analysis of environmental microbiomes. J Microbiol Methods 2023; 214:106841. [PMID: 37832922 DOI: 10.1016/j.mimet.2023.106841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
Quantification of the abundance and understanding of the dynamics of the microbial communities is essential to establish a basis for microbiome characterization. The conventional techniques used for the quantification of microbes are complicated and time-consuming. With scientific advancement, many techniques evolved and came into account. Among them, flow cytometry is a robust, high-throughput technique through which microbial dynamics, morphology, microbial distribution, physiological characteristics, and many more attributes can be studied in a high-throughput manner with comparatively less time and resources. Flow cytometry, when combined with other omics-based methods, offers a rapid and efficient platform to analyze and understand the composition of microbiome at the cellular level. The microbial diversity observed through flow cytometry will not be equivalent to that obtained by sequencing methods, but this integrated approach holds great potential for high throughput characterization of microbiomes. Flow cytometry is regarded as an established characterization tool in haematology, oncology, immunology, and medical microbiology research; however, its application in environmental microbiology is yet to be explored. This comprehensive review aims to delve into the diverse environmental applications of flow cytometry across various domains, including but not limited to bioremediation, landfills, anaerobic digestion, industrial bioprocesses, water quality regulation, and soil quality regulation. By conducting an in-depth analysis, this article seeks to shed light on the potential benefits and challenges associated with the utilization of flow cytometry in addressing environmental concerns.
Collapse
Affiliation(s)
- Madhumita Priyadarsini
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Jeetesh Kushwaha
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Kailash Pati Pandey
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Jyoti Rani
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Abhishek S Dhoble
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
9
|
Moghimi N, Hosseini SA, Dalan AB, Mohammadrezaei D, Goldman A, Kohandel M. Controlled tumor heterogeneity in a co-culture system by 3D bio-printed tumor-on-chip model. Sci Rep 2023; 13:13648. [PMID: 37607994 PMCID: PMC10444838 DOI: 10.1038/s41598-023-40680-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023] Open
Abstract
Cancer treatment resistance is a caused by presence of various types of cells and heterogeneity within the tumor. Tumor cell-cell and cell-microenvironment interactions play a significant role in the tumor progression and invasion, which have important implications for diagnosis, and resistance to chemotherapy. In this study, we develop 3D bioprinted in vitro models of the breast cancer tumor microenvironment made of co-cultured cells distributed in a hydrogel matrix with controlled architecture to model tumor heterogeneity. We hypothesize that the tumor could be represented by a cancer cell-laden co-culture hydrogel construct, whereas its microenvironment can be modeled in a microfluidic chip capable of producing a chemical gradient. Breast cancer cells (MCF7 and MDA-MB-231) and non-tumorigenic mammary epithelial cells (MCF10A) were embedded in the alginate-gelatine hydrogels and printed using a multi-cartridge extrusion bioprinter. Our approach allows for precise control over position and arrangements of cells in a co-culture system, enabling the design of various tumor architectures. We created samples with two different types of cells at specific initial locations, where the density of each cell type was carefully controlled. The cells were either randomly mixed or positioned in sequential layers to create cellular heterogeneity. To study cell migration toward chemoattractant, we developed a chemical microenvironment in a chamber with a gradual chemical gradient. As a proof of concept, we studied different migration patterns of MDA-MB-231 cells toward the epithelial growth factor gradient in presence of MCF10A cells in different ratios using this device. Our approach involves the integration of 3D bioprinting and microfluidic devices to create diverse tumor architectures that are representative of those found in various patients. This provides an excellent tool for studying the behavior of cancer cells with high spatial and temporal resolution.
Collapse
Affiliation(s)
- Nafiseh Moghimi
- Department of Applied Mathematics, University of Waterloo, Waterloo, Canada.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Seied Ali Hosseini
- Electrical Engineering Department, University of Waterloo, Waterloo, Canada
| | - Altay Burak Dalan
- Department of Applied Mathematics, University of Waterloo, Waterloo, Canada
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | | | - Aaron Goldman
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, Canada
| |
Collapse
|
10
|
Lau SY, Kang M, Hisey CL, Chamley LW. Studying exogenous extracellular vesicle biodistribution by in vivo fluorescence microscopy. Dis Model Mech 2023; 16:dmm050074. [PMID: 37526034 PMCID: PMC10417515 DOI: 10.1242/dmm.050074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles released from cells that play a crucial role in many physiological processes and pathological mechanisms. As such, there is great interest in their biodistribution. One currently accessible technology to study their fate in vivo involves fluorescent labelling of exogenous EVs followed by whole-animal imaging. Although this is not a new technology, its translation from studying the fate of whole cells to subcellular EVs requires adaptation of the labelling techniques, excess dye removal and a refined experimental design. In this Review, we detail the methods and considerations for using fluorescence in vivo and ex vivo imaging to study the biodistribution of exogenous EVs and their roles in physiology and disease biology.
Collapse
Affiliation(s)
- Sien Yee Lau
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
| | - Matthew Kang
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
| | - Colin L. Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Lawrence W. Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
11
|
Yaffe ZA, Ding S, Sung K, Chohan V, Marchitto L, Doepker L, Ralph D, Nduati R, Matsen FA, Finzi A, Overbaugh J. Reconstruction of a polyclonal ADCC antibody repertoire from an HIV-1 non-transmitting mother. iScience 2023; 26:106762. [PMID: 37216090 PMCID: PMC10196594 DOI: 10.1016/j.isci.2023.106762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/24/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Human natural history and vaccine studies support a protective role of antibody dependent cellular cytotoxicity (ADCC) activity against many infectious diseases. One setting where this has consistently been observed is in HIV-1 vertical transmission, where passively acquired ADCC activity in HIV-exposed infants has correlated with reduced acquisition risk and reduced pathogenesis in HIV+ infants. However, the characteristics of HIV-specific antibodies comprising a maternal plasma ADCC response are not well understood. Here, we reconstructed monoclonal antibodies (mAbs) from memory B cells from late pregnancy in mother MG540, who did not transmit HIV to her infant despite several high-risk factors. Twenty mAbs representing 14 clonal families were reconstructed, which mediated ADCC and recognized multiple HIV Envelope epitopes. In experiments using Fc-defective variants, only combinations of several mAbs accounted for the majority of plasma ADCC of MG540 and her infant. We present these mAbs as evidence of a polyclonal repertoire with potent HIV-directed ADCC activity.
Collapse
Affiliation(s)
- Zak A. Yaffe
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Shilei Ding
- Centre de Recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Vrasha Chohan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lorie Marchitto
- Centre de Recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Laura Doepker
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Duncan Ralph
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ruth Nduati
- Department of Paediatrics and Child Health, University of Nairobi, Kenyatta National Hospital, Nairobi, Kenya
| | - Frederick A. Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98109, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
12
|
Tippalagama R, Chihab LY, Kearns K, Lewis S, Panda S, Willemsen L, Burel JG, Lindestam Arlehamn CS. Antigen-specificity measurements are the key to understanding T cell responses. Front Immunol 2023; 14:1127470. [PMID: 37122719 PMCID: PMC10140422 DOI: 10.3389/fimmu.2023.1127470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Antigen-specific T cells play a central role in the adaptive immune response and come in a wide range of phenotypes. T cell receptors (TCRs) mediate the antigen-specificities found in T cells. Importantly, high-throughput TCR sequencing provides a fingerprint which allows tracking of specific T cells and their clonal expansion in response to particular antigens. As a result, many studies have leveraged TCR sequencing in an attempt to elucidate the role of antigen-specific T cells in various contexts. Here, we discuss the published approaches to studying antigen-specific T cells and their specific TCR repertoire. Further, we discuss how these methods have been applied to study the TCR repertoire in various diseases in order to characterize the antigen-specific T cells involved in the immune control of disease.
Collapse
|
13
|
Cho J, Min HY, Lee HY. Isolation of slow-cycling cancer cells from lung patient-derived xenograft using carboxyfluorescein-succinimidyl ester retention-mediated cell sorting. STAR Protoc 2023; 4:102167. [PMID: 36924504 PMCID: PMC10026031 DOI: 10.1016/j.xpro.2023.102167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
The slow-cycling subpopulation plays an important role in anticancer drug resistance and tumor recurrence. Here, we describe a clinically relevant patient-derived xenograft model and a carboxyfluorescein succinimidyl ester dye that is diluted in a cell proliferation-dependent manner. We detail steps to separate active-cycling cancer cells and slow-cycling cancer cells (SCCs) in heterogeneous cancer populations to confirm their different cellular properties. This protocol can be used to distinguish SCCs, investigate their biology, and develop strategies for anticancer therapeutics. For complete details on the use and execution of this protocol, please refer to Cho et al. (2021).1.
Collapse
Affiliation(s)
- Jaebeom Cho
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
14
|
Cai F, Ren Y, Dai J, Yang J, Shi X. Effects of Various Cell Surface Engineering Reactions on the Biological Behavior of Mammalian Cells. Macromol Biosci 2023; 23:e2200379. [PMID: 36579789 DOI: 10.1002/mabi.202200379] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Cell surface engineering technologies can regulate cell function and behavior by modifying the cell surface. Previous studies have mainly focused on investigating the effects of cell surface engineering reactions and materials on cell activity. However, they do not comprehensively analyze other cellular processes. This study exploits covalent bonding, hydrophobic interactions, and electrostatic interactions to modify the macromolecules succinimide ester-methoxy polyethylene glycol (NHS-mPEG), distearoyl phosphoethanolamine-methoxy polyethylene glycol (DSPE-mPEG), and poly-L-lysine (PLL), respectively, on the cell surface. This work systematically investigates the effects of the three surface engineering reactions on the behavior of human umbilical vein endothelial cells (HUVECs) and human skin fibroblasts, including viability, growth, proliferation, cell cycle, adhesion, and migration. The results reveals that the PLL modification method notably affects cell viability and G2/M arrest and has a short modification duration. However, the DSPE-mPEG and NHS-mPEG modification methods have little effect on cell viability and proliferation but have a prolonged modification duration. Moreover, the DSPE-mPEG modification method highly affects cell adherence. Further, the NHS-mPEG modification method can significantly improve the migration ability of HUVECs by reducing the area of focal adhesions. The findings of this study will contribute to the application of cell surface engineering technology in the biomedical field.
Collapse
Affiliation(s)
- Fengying Cai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yafeng Ren
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jiajia Dai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| |
Collapse
|
15
|
Magoling BJA, Wu AYT, Chen YJ, Wong WWT, Chuo STY, Huang HC, Sung YC, Hsieh HT, Huang P, Lee KZ, Huang KW, Chen RH, Chen Y, Lai CPK. Membrane Protein Modification Modulates Big and Small Extracellular Vesicle Biodistribution and Tumorigenic Potential in Breast Cancers In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208966. [PMID: 36609913 DOI: 10.1002/adma.202208966] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) are released by cells to mediate intercellular communication under pathological and physiological conditions. While small EVs (sEVs; <100-200 nm, exosomes) are intensely investigated, the properties and functions of medium and large EVs (big EVs (bEVs); >200 nm, microvesicles) are less well explored. Here, bEVs and sEVs are identified as distinct EV populations, and it is determined that bEVs are released in a greater bEV:sEV ratio in the aggressive human triple-negative breast cancer (TNBC) subtype. PalmGRET, bioluminescence-resonance-energy-transfer (BRET)-based EV reporter, reveals dose-dependent EV biodistribution at nonlethal and physiological EV dosages, as compared to lipophilic fluorescent dyes. Remarkably, the bEVs and sEVs exhibit unique biodistribution profiles, yet individually promote in vivo tumor growth in a syngeneic immunocompetent TNBC breast tumor murine model. The bEVs and sEVs share mass-spectrometry-identified tumor-progression-associated EV surface membrane proteins (tpEVSurfMEMs), which include solute carrier family 29 member 1, Cd9, and Cd44. tpEVSurfMEM depletion attenuates EV lung organotropism, alters biodistribution, and reduces protumorigenic potential. This study identifies distinct in vivo property and function of bEVs and sEVs in breast cancer, which suggest the significant role of bEVs in diseases, diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Bryan John Abel Magoling
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
| | - Anthony Yan-Tang Wu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yen-Ju Chen
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Wendy Wan-Ting Wong
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Steven Ting-Yu Chuo
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Hsi-Chien Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hsin Tzu Hsieh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Poya Huang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Kang-Zhang Lee
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Kuan-Wei Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ruey-Hwa Chen
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
16
|
Cotner M, Meng S, Jost T, Gardner A, De Santiago C, Brock A. Integration of quantitative methods and mathematical approaches for the modeling of cancer cell proliferation dynamics. Am J Physiol Cell Physiol 2023; 324:C247-C262. [PMID: 36503241 PMCID: PMC9886359 DOI: 10.1152/ajpcell.00185.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Physiological processes rely on the control of cell proliferation, and the dysregulation of these processes underlies various pathological conditions, including cancer. Mathematical modeling can provide new insights into the complex regulation of cell proliferation dynamics. In this review, we first examine quantitative experimental approaches for measuring cell proliferation dynamics in vitro and compare the various types of data that can be obtained in these settings. We then explore the toolbox of common mathematical modeling frameworks that can describe cell behavior, dynamics, and interactions of proliferation. We discuss how these wet-laboratory studies may be integrated with different mathematical modeling approaches to aid the interpretation of the results and to enable the prediction of cell behaviors, specifically in the context of cancer.
Collapse
Affiliation(s)
- Michael Cotner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Sarah Meng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Tyler Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Andrea Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Carolina De Santiago
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
17
|
Wuttisarnwattana P, Eid S, Wilson DL, Cooke KR. Assessment of therapeutic role of mesenchymal stromal cells in mouse models of graft-versus-host disease using cryo-imaging. Sci Rep 2023; 13:1698. [PMID: 36717650 PMCID: PMC9886911 DOI: 10.1038/s41598-023-28478-3] [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: 06/14/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Insights regarding the biodistribution and homing of mesenchymal stromal cells (MSCs), as well as their interaction with alloreactive T-cells are critical for understanding how MSCs can regulate graft-versus-host disease (GVHD) following allogeneic (allo) bone marrow transplantation (BMT). We developed novel assays based on 3D, microscopic, cryo-imaging of whole-mouse-sized volumes to assess the therapeutic potential of human MSCs using an established mouse GVHD model. Following infusion, we quantitatively tracked fluorescently labeled, donor-derived, T-cells and third party MSCs in BMT recipients using multispectral cryo-imaging. Specific MSC homing sites were identified in the marginal zones in the spleen and the lymph nodes, where we believe MSC immunomodulation takes place. The number of MSCs found in spleen of the allo BMT recipients was about 200% more than that observed in the syngeneic group. To more carefully define the effects MSCs had on T cell activation and expansion, we developed novel T-cell proliferation assays including secondary lymphoid organ (SLO) enlargement and Carboxyfluoescein succinimidyl ester (CFSE) dilution. As anticipated, significant SLO volume enlargement and CFSE dilution was observed in allo but not syn BMT recipients due to rapid proliferation and expansion of labeled T-cells. MSC treatment markedly attenuated CFSE dilution and volume enlargement of SLO. These assays confirm evidence of potent, in vivo, immunomodulatory properties of MSC following allo BMT. Our innovative platform includes novel methods for tracking cells of interest as well as assessing therapeutic function of MSCs during GVHD induction. Our results support the use of MSCs treatment or prevention of GVHD and illuminate the wider adoption of MSCs as a standard medicinal cell therapy.
Collapse
Affiliation(s)
- Patiwet Wuttisarnwattana
- Optimization Theory and Applications for Engineering Systems Research Group, Department of Computer Engineering, Excellence Center in Infrastructure Technology and Transportation Engineering, Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, Thailand.
| | - Saada Eid
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Kenneth R Cooke
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
18
|
Apoptotic extracellular vesicles are metabolized regulators nurturing the skin and hair. Bioact Mater 2023; 19:626-641. [PMID: 35600968 PMCID: PMC9109130 DOI: 10.1016/j.bioactmat.2022.04.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/20/2022] [Accepted: 04/20/2022] [Indexed: 12/24/2022] Open
Abstract
Over 300 billion of cells die every day in the human body, producing a large number of endogenous apoptotic extracellular vesicles (apoEVs). Also, allogenic stem cell transplantation, a commonly used therapeutic approach in current clinical practice, generates exogenous apoEVs. It is well known that phagocytic cells engulf and digest apoEVs to maintain the body's homeostasis. In this study, we show that a fraction of exogenous apoEVs is metabolized in the integumentary skin and hair follicles. Mechanistically, apoEVs activate the Wnt/β-catenin pathway to facilitate their metabolism in a wave-like pattern. The migration of apoEVs is enhanced by treadmill exercise and inhibited by tail suspension, which is associated with the mechanical force-regulated expression of DKK1 in circulation. Furthermore, we show that exogenous apoEVs promote wound healing and hair growth via activation of Wnt/β-catenin pathway in skin and hair follicle mesenchymal stem cells. This study reveals a previously unrecognized metabolic pathway of apoEVs and opens a new avenue for exploring apoEV-based therapy for skin and hair disorders. Exogenous infused apoEVs are partly metabolized from the integumentary skin and hair follicles. ApoEVs activate Wnt/β-catenin pathway to facilitate their elimination in a wave-like pattern. Exercise can enhance apoEV metabolism through Wnt/β-catenin pathway. MSC-derived apoEVs promote wound healing and hair growth.
Collapse
|
19
|
Semenov AN, Gvozdev DA, Moysenovich AM, Zlenko DV, Parshina EY, Baizhumanov AA, Budylin GS, Maksimov EG. Probing Red Blood Cell Membrane Microviscosity Using Fluorescence Anisotropy Decay Curves of the Lipophilic Dye PKH26. Int J Mol Sci 2022; 23:ijms232415767. [PMID: 36555408 PMCID: PMC9781149 DOI: 10.3390/ijms232415767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Red blood cell (RBC) aggregation and deformation are governed by the molecular processes occurring on the membrane. Since several social important diseases are accompanied by alterations in RBC aggregation and deformability, it is important to develop a diagnostic parameter of RBC membrane structural integrity and stability. In this work, we propose membrane microviscosity assessed by time-resolved fluorescence anisotropy of the lipophilic PKH26 fluorescent probe as a diagnostic parameter. We measured the fluorescence decay curves of the PKH26 probe in the RBC membrane to establish the optimal parameters of the developed fluorescence assay. We observed a complex biphasic profile of the fluorescence anisotropy decay characterized by two correlation times corresponding to the rotational diffusion of free PKH26, and membrane-bounded molecules of the probe. The developed assay allowed us to estimate membrane microviscosity ηm in the range of 100-500 cP depending on the temperature, which paves the way for assessing RBC membrane properties in clinical applications as predictors of blood microrheological abnormalities.
Collapse
Affiliation(s)
- Alexey N. Semenov
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Daniil A. Gvozdev
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Anastasia M. Moysenovich
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Dmitry V. Zlenko
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Evgenia Yu. Parshina
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Adil A. Baizhumanov
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory Str., 119991 Moscow, Russia
| | - Gleb S. Budylin
- Laboratory of Clinical Biophotonics, Biomedical Science and Technology Park, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Eugene G. Maksimov
- Interdisciplinary Scientific and Educational School, Molecular Technologies of the Living Systems and Synthetic Biology, M.V. Lomonosov Moscow State University, 1 Leninskie Gory Str., 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
20
|
Chain-End Functionalization of Poly(ε-caprolactone) for Chemical Binding with Gelatin: Binary Electrospun Scaffolds with Improved Physico-Mechanical Characteristics and Cell Adhesive Properties. Polymers (Basel) 2022; 14:polym14194203. [PMID: 36236153 PMCID: PMC9570970 DOI: 10.3390/polym14194203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Composite biocompatible scaffolds, obtained using the electrospinning (ES) technique, are highly promising for biomedical application thanks to their high surface area, porosity, adjustable fiber diameter, and permeability. However, the combination of synthetic biodegradable (such as poly(ε-caprolactone) PCL) and natural (such as gelatin Gt) polymers is complicated by the problem of low compatibility of the components. Previously, this problem was solved by PCL grafting and/or Gt cross-linking after ES molding. In the present study, composite fibrous scaffolds consisting of PCL and Gt were fabricated by the electrospinning (ES) method using non-functionalized PCL1 or NHS-functionalized PCL2 and hexafluoroisopropanol as a solvent. To provide covalent binding between PCL2 and Gt macromolecules, NHS-functionalized methyl glutarate was synthesized and studied in model reactions with components of spinning solution. It was found that selective formation of amide bonds, which provide complete covalent bonding of Gt in PCL/Gt composite, requires the presence of weak acid. With the use of the optimized ES method, fibrous mats with different PCL/Gt ratios were prepared. The sample morphology (SEM), hydrolytic resistance (FT-IR), cell adhesion and viability (MTT assay), cell penetration (fluorescent microscopy), and mechanical characteristics of the samples were studied. PCL2-based films with a Gt content of 20 wt% have demonstrated the best set of properties.
Collapse
|
21
|
Salman MI, Al-Shammari AM, Emran MA. 3-Dimensional coculture of breast cancer cell lines with adipose tissue–Derived stem cells reveals the efficiency of oncolytic Newcastle disease virus infection via labeling technology. Front Mol Biosci 2022; 9:754100. [PMID: 36172043 PMCID: PMC9511405 DOI: 10.3389/fmolb.2022.754100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Abstract
Oncolytic virotherapy is one of the emerging biological therapeutics that needs a more efficient in vitro tumor model to overcome the two-dimensional (2D) monolayer tumor cell culture model’s inability to maintain tissue-specific structure. This is to offer significant prognostic preclinical assessment findings. One of the best models that can mimic the in vivo model in vitro are the three-dimensional (3D) tumor–normal cell coculture systems, which can be employed in preclinical oncolytic virus therapeutics. Thus, we developed our 3D coculture system in vitro using two types of breast cancer cell lines showing different receptor statuses cocultured with adipose tissue–derived mesenchymal stem cells. The cells were cultured in a floater tissue culture plate to allow spheroids formation, and then the spheroids were collected and transferred to a scaffold spheroids dish. These 3D culture systems were used to evaluate oncolytic Newcastle disease virus AMHA1 strain infectivity and antitumor activity using a tracking system of the Newcastle disease virus (NDV) labeled with fluorescent PKH67 linker to follow the virus entry into target cells. This provides evidence that the NDV AMHA1 strain is an efficient oncolytic agent. The fluorescently detected virus particles showed high intensity in both coculture spheres. Strategies for chemically introducing fluorescent dyes into NDV particles extract quantitative information from the infected cancer models. In conclusion, the results indicate that the NDV AMHA1 strain efficiently replicates and induces an antitumor effect in cancer–normal 3D coculture systems, indicating efficient clinical outcomes.
Collapse
Affiliation(s)
- Marwa Ibrahim Salman
- Department of Biotechnology, College of Science, University of Baghdad, Baghdad, Iraq
| | - Ahmed Majeed Al-Shammari
- Department of Experimental Therapy, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
- *Correspondence: Ahmed Majeed Al-Shammari,
| | - Mahfodha Abbas Emran
- Department of Biotechnology, College of Science, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
22
|
Liu H, Liu S, Xiao Y, Song W, Li H, Ho LWC, Shen Z, Choi CHJ. A pH-Reversible Fluorescent Probe for in Situ Imaging of Extracellular Vesicles and Their Secretion from Living Cells. NANO LETTERS 2021; 21:9224-9232. [PMID: 34724785 DOI: 10.1021/acs.nanolett.1c03110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Our knowledge in how extracellular vesicles (EVs) are secreted from cells remains inadequate due to the limited technologies available for visualizing them in situ. We report a pH-reversible boron dipyrromethene (BODIPY) fluorescent probe for confocal imaging of EVs secreted from living cells without inducing severe cytotoxicity. This probe predominantly assumes a non-fluorescent leuco-BODIPY form under basic conditions, but it gradually switches to its fluorescent parent BODIPY form upon acidification; such pH transition empowers the imaging of acidic EVs (such as CD81-enriched exosomes and extracellular multivesicular bodies) in weakly basic culture medium and intracellular acidic precursor EVs in weakly basic cytoplasm, with minimal false positive signals frequently encountered for "always-on" dyes. Joint application of this probe with plasmid transfection reveals the secretion of some EVs from cellular pseudopodia via microtubule trackways. This probe may provide mechanistic insights into the extracellular transport of EVs and support the development of EV-based nanomedicines.
Collapse
Affiliation(s)
- Hanzhuang Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Shaorui Liu
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Yu Xiao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Wenting Song
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huize Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Lok Wai Cola Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Zhen Shen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| |
Collapse
|
23
|
Abstract
Cell cycle involves a series of changes that lead to cell growth and division. Cell cycle analysis is crucial to understand cellular responses to changing environmental conditions. Since its inception, flow cytometry has been particularly useful for cell cycle analysis at single cell level due to its speed and precision. Previously, flow cytometric cell cycle analysis relied solely on the measurement of cellular DNA content. Later, methods were developed for multiparametric analysis. This review explains the journey of flow cytometry to understand different molecular and cellular events underlying cell cycle using various protocols. Recent advances in the field that overcome the shortcomings of traditional flow cytometry and expand its scope for cell cycle studies are also discussed.
Collapse
|
24
|
Kasten-Jolly J, Lawrence DA. In Vitro Evaluation of Toxicant Influences on the Immune System. ACTA ACUST UNITED AC 2021; 84:e95. [PMID: 32539242 DOI: 10.1002/cptx.95] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Culture of human peripheral blood mononuclear cells (PBMCs) still remains a convenient and sensitive method for measurement of a person's immune system health. Basic elements of the process, namely PBMC purification and culture medium formulation, were first reported in the late 1960s, and the utility of the method for clinical application was reported in the 1970s. Clinically, the approach can provide information about the ability of an individual's immune system to fight off attacks by various pathogens. Over the years, the method has undergone many improvements, which have been aided by advancements made in flow cytometry technology and the development of fluorescent reagents. The protocols presented here describe flow cytometry-based techniques for PBMC culture that can be employed to determine the impact of various environmental toxicants on the immune system. A major advantage of these procedures is that they will provide information about a toxicant or drug through in vitro methods. As the relationship between exposures to certain toxicants and an individual's response to vaccinations has been of concern, one of the protocols described shows how to test an environmental toxicant for potential modification of the immune response to vaccine antigen(s). © 2020 Wiley Periodicals LLC. Basic Protocol 1: Measurement of cell proliferation Support Protocol 1: Blood cell counting Support Protocol 2: Measurement of cell viability after culturing Basic Protocol 2: Identification of affected naïve/memory cell subsets.
Collapse
Affiliation(s)
- Jane Kasten-Jolly
- Wadsworth Center, New York State Department of Health, Albany, New York
| | - David A Lawrence
- Wadsworth Center, New York State Department of Health, Albany, New York.,Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, New York
| |
Collapse
|
25
|
Tick extracellular vesicles enable arthropod feeding and promote distinct outcomes of bacterial infection. Nat Commun 2021; 12:3696. [PMID: 34140472 PMCID: PMC8211691 DOI: 10.1038/s41467-021-23900-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles are thought to facilitate pathogen transmission from arthropods to humans and other animals. Here, we reveal that pathogen spreading from arthropods to the mammalian host is multifaceted. Extracellular vesicles from Ixodes scapularis enable tick feeding and promote infection of the mildly virulent rickettsial agent Anaplasma phagocytophilum through the SNARE proteins Vamp33 and Synaptobrevin 2 and dendritic epidermal T cells. However, extracellular vesicles from the tick Dermacentor andersoni mitigate microbial spreading caused by the lethal pathogen Francisella tularensis. Collectively, we establish that tick extracellular vesicles foster distinct outcomes of bacterial infection and assist in vector feeding by acting on skin immunity. Thus, the biology of arthropods should be taken into consideration when developing strategies to control vector-borne diseases.
Collapse
|
26
|
Exploring interactions between extracellular vesicles and cells for innovative drug delivery system design. Adv Drug Deliv Rev 2021; 173:252-278. [PMID: 33798644 DOI: 10.1016/j.addr.2021.03.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are submicron cell-secreted structures containing proteins, nucleic acids and lipids. EVs can functionally transfer these cargoes from one cell to another to modulate physiological and pathological processes. Due to their presumed biocompatibility and capacity to circumvent canonical delivery barriers encountered by synthetic drug delivery systems, EVs have attracted considerable interest as drug delivery vehicles. However, it is unclear which mechanisms and molecules orchestrate EV-mediated cargo delivery to recipient cells. Here, we review how EV properties have been exploited to improve the efficacy of small molecule drugs. Furthermore, we explore which EV surface molecules could be directly or indirectly involved in EV-mediated cargo transfer to recipient cells and discuss the cellular reporter systems with which such transfer can be studied. Finally, we elaborate on currently identified cellular processes involved in EV cargo delivery. Through these topics, we provide insights in critical effectors in the EV-cell interface which may be exploited in nature-inspired drug delivery strategies.
Collapse
|
27
|
Slow-cycling (dormant) cancer cells in therapy resistance, cancer relapse and metastasis. Semin Cancer Biol 2021; 78:90-103. [PMID: 33979674 DOI: 10.1016/j.semcancer.2021.04.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that cancer cell heterogeneity and plasticity constitute major barriers to effective clinical treatments and long-term therapeutic efficacy. Research in the past two decades suggest that virtually all treatment-naive human cancers harbor subsets of cancer cells that possess many of the cardinal features of normal stem cells. Such stem-like cancer cells, operationally defined as cancer stem cells (CSCs), are frequently quiescent and dynamically change and evolve during tumor progression and therapeutic interventions. Intrinsic tumor cell heterogeneity is reflected in a different aspect in that tumors also harbor a population of slow-cycling cells (SCCs) that are not in the proliferative cell cycle and thus are intrinsically refractory to anti-mitotic drugs. In this Perspective, we focus our discussions on SCCs in cancer and on various methodologies that can be employed to enrich and purify SCCs, compare the similarities and differences between SCCs, CSCs and cancer cells undergoing EMT, and present evidence for the involvement of SCCs in surviving anti-neoplastic treatments, mediating tumor relapse, maintaining tumor dormancy and mediating metastatic dissemination. Our discussions make it clear that an in-depth understanding of the biological properties of SCCs in cancer will be instrumental to developing new therapeutic strategies to prevent tumor relapse and distant metastasis.
Collapse
|
28
|
Cryopreservation and Transplantation of Spermatogonial Stem Cells. Methods Mol Biol 2021. [PMID: 33606221 DOI: 10.1007/978-1-0716-0970-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Cryopreservation as a method that enables long-term storage of biological material has long been used for the conservation of valuable zebrafish genetic resources. However, currently, only spermatozoa of zebrafish can be successfully cryopreserved, while protocols for cryopreservation of eggs and embryos have not yet been fully developed. Transplantation of germline stem cells (GSCs) has risen as a favorable method that can bypass the current problem in cryopreservation of female genetic resources and can lead to reconstitution of fish species and lines through surrogate production. Here, we describe essential steps needed for the cryopreservation of spermatogonial stem cells (SSCs) and their utilization in the conservation of zebrafish genetic resources through SSC transplantation and surrogate production.
Collapse
|
29
|
Shimomura T, Seino R, Umezaki K, Shimoda A, Ezoe T, Ishiyama M, Akiyoshi K. New Lipophilic Fluorescent Dyes for Labeling Extracellular Vesicles: Characterization and Monitoring of Cellular Uptake. Bioconjug Chem 2021; 32:680-684. [PMID: 33719402 DOI: 10.1021/acs.bioconjchem.1c00068] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PKH dyes, which are currently the most widely used fluorescent probes for extracellular vesicle (EV) labeling, have some limitations. For example, these dyes tend to aggregate, leading to formation of EV-like nanoparticles that can be taken up by cells. Moreover, it has been suggested that PKH dyes trigger an enlargement of EVs because of membrane fusion or intercalation. To overcome these limitations, we developed three novel extracellular vesicular-membrane-binding fluorescent probes-Mem dye-Green, Mem dye-Red, and Mem dye-Deep Red-for monitoring EV uptake into cells. The dyes contain a cyanine group as a fluorescent scaffold and amphiphilic moieties on the cyanine. The three dyes have different photophysical characteristics. To investigate the characteristics of the Mem dyes for EV labeling, we performed nanoparticle tracking, zeta potential measurements, and confocal microscopy. The dyes enable highly sensitive fluorescence imaging of EVs. They can also be used to observe EV dynamics in live cells. The Mem dyes show excellent EV labeling with no aggregation and less particle enlargement.
Collapse
Affiliation(s)
- Takashi Shimomura
- Dojindo Laboratories, 2025-5 Tabaru, Mashiki-machi, Kumamoto 861-2202, Japan
| | - Ryo Seino
- Dojindo Laboratories, 2025-5 Tabaru, Mashiki-machi, Kumamoto 861-2202, Japan
| | - Kaori Umezaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Asako Shimoda
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Takatoshi Ezoe
- Dojindo Laboratories, 2025-5 Tabaru, Mashiki-machi, Kumamoto 861-2202, Japan
| | - Munetaka Ishiyama
- Dojindo Laboratories, 2025-5 Tabaru, Mashiki-machi, Kumamoto 861-2202, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
30
|
Determination of Cytotoxic Activities Against Melanoma Cells Using Flow Cytometry. Methods Mol Biol 2021. [PMID: 33704709 DOI: 10.1007/978-1-0716-1205-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Within the adaptive and innate immune system, effector lymphocytes known as cytotoxic T cells (CTLs) or natural killer (NK) cells play an essential role in host defense against tumor cells and virally infected cells. Here we describe a flow cytometry-based method to quantify CTLs or NK cell cytotoxic activity against melanoma cells. In this assay, spleen cells, peripheral blood mononuclear cells (PBMCs), or purified NK cell preparations are co-incubated at different ratios with a target tumor cell line. The target cells are pre-labeled with a fluorescent dye to allow their discrimination from the effector cells. After the incubation period, killed target cells are identified by a nucleic acid stain, which specifically permeates dead cells. This method is amenable to both diagnostic and research applications.
Collapse
|
31
|
Cho J, Min HY, Lee HJ, Hyun SY, Sim JY, Noh M, Hwang SJ, Park SH, Boo HJ, Lee HJ, Hong S, Park RW, Shin YK, Hung MC, Lee HY. RGS2-mediated translational control mediates cancer cell dormancy and tumor relapse. J Clin Invest 2021; 131:136779. [PMID: 33393490 PMCID: PMC7773398 DOI: 10.1172/jci136779] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Slow-cycling/dormant cancer cells (SCCs) have pivotal roles in driving cancer relapse and drug resistance. A mechanistic explanation for cancer cell dormancy and therapeutic strategies targeting SCCs are necessary to improve patient prognosis, but are limited because of technical challenges to obtaining SCCs. Here, by applying proliferation-sensitive dyes and chemotherapeutics to non-small cell lung cancer (NSCLC) cell lines and patient-derived xenografts, we identified a distinct SCC subpopulation that resembled SCCs in patient tumors. These SCCs displayed major dormancy-like phenotypes and high survival capacity under hostile microenvironments through transcriptional upregulation of regulator of G protein signaling 2 (RGS2). Database analysis revealed RGS2 as a biomarker of retarded proliferation and poor prognosis in NSCLC. We showed that RGS2 caused prolonged translational arrest in SCCs through persistent eukaryotic initiation factor 2 (eIF2α) phosphorylation via proteasome-mediated degradation of activating transcription factor 4 (ATF4). Translational activation through RGS2 antagonism or the use of phosphodiesterase 5 inhibitors, including sildenafil (Viagra), promoted ER stress-induced apoptosis in SCCs in vitro and in vivo under stressed conditions, such as those induced by chemotherapy. Our results suggest that a low-dose chemotherapy and translation-instigating pharmacological intervention in combination is an effective strategy to prevent tumor progression in NSCLC patients after rigorous chemotherapy.
Collapse
Affiliation(s)
- Jaebeom Cho
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Ho Jin Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Seung Yeob Hyun
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Jeong Yeon Sim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Myungkyung Noh
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Su Jung Hwang
- College of Pharmacy, Inje University, Gimhae, Gyungnam, Republic of Korea
| | - Shin-Hyung Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Hye-Jin Boo
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Hyo-Jong Lee
- College of Pharmacy, Inje University, Gimhae, Gyungnam, Republic of Korea
| | - Sungyoul Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| |
Collapse
|
32
|
Rai A, Fang H, Fatmous M, Claridge B, Poh QH, Simpson RJ, Greening DW. A Protocol for Isolation, Purification, Characterization, and Functional Dissection of Exosomes. Methods Mol Biol 2021; 2261:105-149. [PMID: 33420988 DOI: 10.1007/978-1-0716-1186-9_9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Extracellular vesicles (EVs) are membrane-enclosed vesicles released by cells. They carry proteins, nucleic acids, and metabolites which can be transferred to a recipient cell, locally or at a distance, to elicit a functional response. Since their discovery over 30 years ago, the functional repertoire of EVs in both physiological (e.g., organ morphogenesis, embryo implantation) and pathological (e.g., cancer, neurodegeneration) conditions has cemented their crucial role in intercellular communication. Moreover, because the cargo encapsulated within circulating EVs remains protected from degradation, their diagnostic as well as therapeutic (such as drug delivery tool) applications have garnered vested interest. Global efforts have been made to purify EV subtypes from biological fluids and in vitro cell culture media using a variety of strategies and techniques, with a major focus on EVs of endocytic origin called exosomes (30-150 nm in size). Given that the secretome comprises of soluble secreted proteins, protein aggregates, RNA granules, and EV subtypes (such as exosomes, shed microvesicles, apoptotic bodies), it is imperative to purify exosomes to homogeneity if we are to perform biochemical and biophysical characterization and, importantly, functional dissection. Besides understanding the composition of EV subtypes, defining molecular bias of how they reprogram target cells also remains of paramount importance in this area of active research. Here, we outline a systematic "how to" protocol (along with useful insights/tips) to obtain highly purified exosomes and perform their biophysical and biochemical characterization. This protocol employs a mass spectrometry-based proteomics approach to characterize the protein composition of exosomes. We also provide insights on different isolation strategies and their usefulness in various downstream applications. We outline protocols for lipophilic labeling of exosomes to study uptake by a recipient cell, investigating cellular reprogramming using proteomics and studying functional response to exosomes in the Transwell-Matrigel™ Invasion assay.
Collapse
Affiliation(s)
- Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Monique Fatmous
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Bethany Claridge
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Niedermair T, Lukas C, Li S, Stöckl S, Craiovan B, Brochhausen C, Federlin M, Herrmann M, Grässel S. Influence of Extracellular Vesicles Isolated From Osteoblasts of Patients With Cox-Arthrosis and/or Osteoporosis on Metabolism and Osteogenic Differentiation of BMSCs. Front Bioeng Biotechnol 2020; 8:615520. [PMID: 33425878 PMCID: PMC7785908 DOI: 10.3389/fbioe.2020.615520] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Studies with extracellular vesicles (EVs), including exosomes, isolated from mesenchymal stem cells (MSC) indicate benefits for the treatment of musculoskeletal pathologies as osteoarthritis (OA) and osteoporosis (OP). However, little is known about intercellular effects of EVs derived from pathologically altered cells that might influence the outcome by counteracting effects from “healthy” MSC derived EVs. We hypothesize, that EVs isolated from osteoblasts of patients with hip OA (coxarthrosis/CA), osteoporosis (OP), or a combination of both (CA/OP) might negatively affect metabolism and osteogenic differentiation of bone-marrow derived (B)MSCs. Methods: Osteoblasts, isolated from bone explants of CA, OP, and CA/OP patients, were compared regarding growth, viability, and osteogenic differentiation capacity. Structural features of bone explants were analyzed via μCT. EVs were isolated from supernatant of naïve BMSCs and CA, OP, and CA/OP osteoblasts (osteogenic culture for 35 days). BMSC cultures were stimulated with EVs and subsequently, cell metabolism, osteogenic marker gene expression, and osteogenic differentiation were analyzed. Results: Trabecular bone structure was different between the three groups with lowest number and highest separation in the CA/OP group. Viability and Alizarin red staining increased over culture time in CA/OP osteoblasts whereas growth of osteoblasts was comparable. Alizarin red staining was by trend higher in CA compared to OP osteoblasts after 35 days and ALP activity was higher after 28 and 35 days. Stimulation of BMSC cultures with CA, OP, and CA/OP EVs did not affect proliferation but increased caspase 3/7-activity compared to unstimulated BMSCs. BMSC viability was reduced after stimulation with CA and CA/OP EVs compared to unstimulated BMSCs or stimulation with OP EVs. ALP gene expression and activity were reduced in BMSCs after stimulation with CA, OP, and CA/OP EVs. Stimulation of BMSCs with CA EVs reduced Alizarin Red staining by trend. Conclusion: Stimulation of BMSCs with EVs isolated from CA, OP, and CA/OP osteoblasts had mostly catabolic effects on cell metabolism and osteogenic differentiation irrespective of donor pathology and reflect the impact of tissue microenvironment on cell metabolism. These catabolic effects are important for understanding differences in effects of EVs on target tissues/cells when harnessing them as therapeutic drugs.
Collapse
Affiliation(s)
- Tanja Niedermair
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Christoph Lukas
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Shushan Li
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Sabine Stöckl
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Benjamin Craiovan
- Chair of Arthroplasty, Center for Orthopaedics and Trauma Surgery, University Hospital Giessen and Marburg GmbH, Marburg, Germany
| | | | - Marianne Federlin
- Department of Conservative Dentistry and Periodontology, University Medical Center Regensburg, Regensburg, Germany
| | - Marietta Herrmann
- IInterdisciplinary Center for Clinical Research (IZKF), Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg and Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| |
Collapse
|
34
|
Di Blasi D, Claessen I, Turksma AW, van Beek J, Ten Brinke A. Guidelines for analysis of low-frequency antigen-specific T cell results: Dye-based proliferation assay vs 3H-thymidine incorporation. J Immunol Methods 2020; 487:112907. [PMID: 33152332 DOI: 10.1016/j.jim.2020.112907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
It is generally recognized that dysregulation of the immune system plays a critical role in many diseases, including autoimmune diseases and cancer. T cells play a crucial role in maintaining self-tolerance, while loss of immune tolerance and T cell activation can lead to severe inflammation and tissue damage. T cell responses have a key role in the effectiveness of vaccination strategies and immunomodulating therapies. Immunomonitoring methods have the ability to elucidate immunological processes, monitor the development of disease and assess therapeutic effects. In this respect, it is of particular interest to evaluate antigen (Ag)-specific T cells by determining their frequency, type and functionality in cellular assays. Nevertheless, Ag-specific T cells are detected infrequently in most diseases using current techniques. Many efforts have been made to develop more sensitive, reproducible, and reliable methods for Ag-specific T cell detection. It has been found that analysis of cellular proliferation can be a useful tool to determine the presence and frequency of Ag-specific T cell and to provides insight into modulation of the T cell response by a specific antigen or therapy. However, the selection of a cut-off value for a positive response and therefore a more accurate interpretation of the data, continues to be a major concern. Here, we provide guidelines to select a proper cut-off for monitoring of Ag-specific CD4+ T cell responses. In vitro Ag-stimulation has been assessed with two methods; a dye-based proliferation assay and 3H-thymidine-based assay. Two cut-off approaches are compared; mean and variance of control wells, and the stimulation index. By evaluating the proliferative response to the in vitro Ag-stimulation using these two methods, we demonstrate the importance of taking into consideration the variability of the control wells to distinguish a positive from a false positive response.
Collapse
Affiliation(s)
- Daniela Di Blasi
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| | - Iris Claessen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands; Sanquin Diagnostics B.V., Amsterdam, the Netherlands
| | | | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| |
Collapse
|
35
|
Exploiting the Natural Properties of Extracellular Vesicles in Targeted Delivery towards Specific Cells and Tissues. Pharmaceutics 2020; 12:pharmaceutics12111022. [PMID: 33114492 PMCID: PMC7692617 DOI: 10.3390/pharmaceutics12111022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication that participate in many physiological/pathological processes. As such, EVs have unique properties related to their origin, which can be exploited for drug delivery applications in cell regeneration, immunosuppression, inflammation, cancer treatment or cardioprotection. Moreover, their cell-like membrane organization facilitates uptake and accumulation in specific tissues and organs, which can be exploited to improve selectivity of cargo delivery. The combination of these properties with the inclusion of drugs or imaging agents can significantly improve therapeutic efficacy and selectivity, reduce the undesirable side effects of drugs or permit earlier diagnosis of diseases. In this review, we will describe the natural properties of EVs isolated from different cell sources and discuss strategies that can be applied to increase the efficacy of targeting drugs or other contents to specific locations. The potential risks associated with the use of EVs will also be addressed.
Collapse
|
36
|
Predicting in vivo therapeutic efficacy of bioorthogonally labeled endothelial progenitor cells in hind limb ischemia models via non-invasive fluorescence molecular tomography. Biomaterials 2020; 266:120472. [PMID: 33120201 DOI: 10.1016/j.biomaterials.2020.120472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/04/2020] [Accepted: 10/18/2020] [Indexed: 01/15/2023]
Abstract
Human embryonic stem cells-derived endothelial progenitor cells (hEPCs) were utilized as cell therapeutics for the treatment of ischemic diseases. However, in vivo tracking of hEPCs for predicting their therapeutic efficacy is very difficult. Herein, we developed bioorthogonal labeling strategy of hEPCs that could non-invasively track them after transplantation in hind limb ischemia models. First, hEPCs were treated with tetraacylated N-azidomannosamine (Ac4ManNAz) for generating unnatural azide groups on the hEPCs surface. Second, near-infrared fluorescence (NIRF) dye, Cy5, conjugated dibenzocylooctyne (DBCO-Cy5) was chemically conjugated to the azide groups on the hEPC surface via copper-free click chemistry, resulting Cy5-hEPCs. The bioorthogonally labeled Cy5-hEPCs showed strong NIRF signal without cytotoxicity and functional perturbation in tubular formation, oxygen consumption and paracrine effect of hEPCs in vitro. In hind limb ischemia models, the distribution and migration of transplanted Cy5-hEPCs were successfully monitored via fluorescence molecular tomography (FMT) for 28 days. Notably, blood reperfusion and therapeutic neovascularization effects were significantly correlated with the initial transplantation forms of Cy5-hEPCs such as 'condensed round shape' and 'spread shape' in the ischemic lesion. The condensed transplanted Cy5-hEPCs substantially increased the therapeutic efficacy of hind limb ischemia, compared to that of spread Cy5-hEPCs. Therefore, our new stem cell labeling strategy can be used to predict therapeutic efficacy in hind limb ischemia and it can be applied a potential application in developing cell therapeutics for regenerative medicine.
Collapse
|
37
|
Experimental limitations of extracellular vesicle-based therapies for the treatment of myocardial infarction. Trends Cardiovasc Med 2020; 31:405-415. [PMID: 32822840 PMCID: PMC8501308 DOI: 10.1016/j.tcm.2020.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are particles secreted by a vast variety of cells and are often recognised to mimic the properties of their parent cell, as such those derived from developmental sources hold promise for the treatment of various diseases including myocardial infarction (MI). Here we review the experimental approaches taken for assessing the therapeutic efficacy of EVs for MI and find overt shortcomings regarding purity of isolated EVs, quantitation, dosing, EV labelling/uptake, route of administration and use of appropriate controls that renders much of the data uninterpretable. Overall, the EV/MI field has suffered from experimental approaches that are not fully standardised or validated. Fundamental improvements in EV study design are required to improve interpretation of efficacy and to ensure reproducibility and comparability across preclinical MI studies.
Collapse
|
38
|
Kundu K, Weber N, Griebler C, Elsner M. Phenotypic heterogeneity as key factor for growth and survival under oligotrophic conditions. Environ Microbiol 2020; 22:3339-3356. [PMID: 32500958 DOI: 10.1111/1462-2920.15106] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 04/09/2020] [Accepted: 05/28/2020] [Indexed: 11/26/2022]
Abstract
Productivity-poor oligotrophic environments are plentiful on earth. Yet it is not well understood how organisms maintain population sizes under these extreme conditions. Most scenarios consider the adaptation of a single microorganism (isogenic) at the cellular level, which increases their fitness in such an environment. However, in oligotrophic environments, the adaptation of microorganisms at population level - that is, the ability of living cells to differentiate into subtypes with specialized attributes leading to the coexistence of different phenotypes in isogenic populations - remains a little-explored area of microbiology research. In this study, we performed experiments to demonstrate that an isogenic population differentiated to two subpopulations under low energy-flux in chemostats. Fluorescence cytometry and turnover rates revealed that these subpopulations differ in their nucleic acid content and metabolic activity. A mechanistic modelling framework for the dynamic adaptation of microorganisms with the consideration of their ability to switch between different phenotypes was experimentally calibrated and validated. Simulation of hypothetical scenarios suggests that responsive diversification upon a change in energy availability offers a competitive advantage over homogenous adaptation for maintaining viability and metabolic activity with time.
Collapse
Affiliation(s)
- Kankana Kundu
- Institute of Groundwater Ecology, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, 85764 Neuherberg, Bavaria, Germany.,Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, Ghent 9000, Belgium
| | - Nina Weber
- Institute of Groundwater Ecology, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, 85764 Neuherberg, Bavaria, Germany
| | - Christian Griebler
- Institute of Groundwater Ecology, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, 85764 Neuherberg, Bavaria, Germany.,Division of Limnology, University of Vienna, Department of Functional and Evolutionary Ecology, Althanstrasse 14, Vienna, 1090, Austria
| | - Martin Elsner
- Institute of Groundwater Ecology, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, 85764 Neuherberg, Bavaria, Germany.,Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Marchioninistrasse 17, D-81377 Munich, Germany
| |
Collapse
|
39
|
Dehghani M, Gulvin SM, Flax J, Gaborski TR. Systematic Evaluation of PKH Labelling on Extracellular Vesicle Size by Nanoparticle Tracking Analysis. Sci Rep 2020; 10:9533. [PMID: 32533028 PMCID: PMC7293335 DOI: 10.1038/s41598-020-66434-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles secreted by cells and can modulate biological activities by transferring their content following uptake into recipient cells. Labelling of EVs is a commonly used technique for understanding their cellular targeting and biodistribution. A reliable fluorescent technique needs to preserve the size of EVs since changes in size may alter their uptake and biodistribution. Lipophilic fluorescent dye molecules such as the PKH family have been widely used for EV labelling. Here, the effect of PKH labelling on the size of EVs was systematically evaluated using nanoparticle tracking analysis (NTA), which is a widely used technique for determining the size and concentration of nanoparticles. NTA analysis showed a size increase in all the PKH labelling conditions tested. As opposed to lipophilic dye molecules, no significant shift in the size of labelled EVs was detected with luminal binding dye molecules such as 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester (CFDA-SE, hereinafter CFSE). This finding suggests that PKH labelling may not be a reliable technique for the tracking of EVs.
Collapse
Affiliation(s)
- Mehdi Dehghani
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, United States
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Shannon M Gulvin
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Jonathan Flax
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Thomas R Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States.
| |
Collapse
|
40
|
Bolat ZB, Islek Z, Demir BN, Yilmaz EN, Sahin F, Ucisik MH. Curcumin- and Piperine-Loaded Emulsomes as Combinational Treatment Approach Enhance the Anticancer Activity of Curcumin on HCT116 Colorectal Cancer Model. Front Bioeng Biotechnol 2020; 8:50. [PMID: 32117930 PMCID: PMC7026030 DOI: 10.3389/fbioe.2020.00050] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/22/2020] [Indexed: 12/24/2022] Open
Abstract
Combination chemotherapy, administrating two chemotherapeutic agents concurrently, comes into prominence, as the heterogeneity or the level of the disease necessitates a collaborative action. Curcumin, isolated from turmeric, and piperine, isolated from black long pepper, are two dietary polyphenols studied for their intrinsic anti-cancer properties against various cancer types including colorectal cancer (CRC). Furthermore, piperine improves the therapeutic effect of curcumin. Addressing this mutual behavior, this study combines curcumin and piperine within emulsome nanoformulations. Curcumin- (CurcuEmulsomes) and piperine-loaded emulsomes (PiperineEmulsomes) have established a uniform, stable, spherical dispersion with average diameters of 184.21 and 248.76 nm, respectively. The solid tripalmitin inner core achieved encapsulation capacities of up to 0.10 mg/ml curcumin and 0.09 mg/ml piperine content. While piperine treatment alone - in its both free and emulsome forms - showed no inhibition in the proliferation of HCT116 cells in vitro, its presence as the second drug agent enhanced curcumin's effect. Combination of 7 μM PiperineEmulsome and 25 μM CurcuEmulsome concentrations was found to be most effective with an inhibition of cell proliferation of about 50% viability. Cell cycle arrest at G2/M phase and induced apoptosis verified the improved anti-cancer characteristics of the therapy. While CurcuEmulsomes achieved a fourfold increase in Caspase 3 level, combination of treatment with PiperineEulsomes achieved a sixfold increase in the level of this apoptotic marker. Combinational treatment of HCT116 cells with CurcuEmulsomes and PiperineEmulsomes improved the anticancer activity of the compounds and highlighted the potential of the approach for further in vivo studies.
Collapse
Affiliation(s)
- Zeynep Busra Bolat
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Zeynep Islek
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Bilun Nas Demir
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Elif Nur Yilmaz
- Graduate School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medicine Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Mehmet Hikmet Ucisik
- Regenerative and Restorative Medicine Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey.,Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
41
|
Lara P, Palma-Florez S, Salas-Huenuleo E, Polakovicova I, Guerrero S, Lobos-Gonzalez L, Campos A, Muñoz L, Jorquera-Cordero C, Varas-Godoy M, Cancino J, Arias E, Villegas J, Cruz LJ, Albericio F, Araya E, Corvalan AH, Quest AFG, Kogan MJ. Gold nanoparticle based double-labeling of melanoma extracellular vesicles to determine the specificity of uptake by cells and preferential accumulation in small metastatic lung tumors. J Nanobiotechnology 2020; 18:20. [PMID: 31973696 PMCID: PMC6979068 DOI: 10.1186/s12951-020-0573-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Extracellular vesicles (EVs) have shown great potential for targeted therapy, as they have a natural ability to pass through biological barriers and, depending on their origin, can preferentially accumulate at defined sites, including tumors. Analyzing the potential of EVs to target specific cells remains challenging, considering the unspecific binding of lipophilic tracers to other proteins, the limitations of fluorescence for deep tissue imaging and the effect of external labeling strategies on their natural tropism. In this work, we determined the cell-type specific tropism of B16F10-EVs towards cancer cell and metastatic tumors by using fluorescence analysis and quantitative gold labeling measurements. Surface functionalization of plasmonic gold nanoparticles was used to promote indirect labeling of EVs without affecting size distribution, polydispersity, surface charge, protein markers, cell uptake or in vivo biodistribution. Double-labeled EVs with gold and fluorescent dyes were injected into animals developing metastatic lung nodules and analyzed by fluorescence/computer tomography imaging, quantitative neutron activation analysis and gold-enhanced optical microscopy. Results We determined that B16F10 cells preferentially take up their own EVs, when compared with colon adenocarcinoma, macrophage and kidney cell-derived EVs. In addition, we were able to detect the preferential accumulation of B16F10 EVs in small metastatic tumors located in lungs when compared with the rest of the organs, as well as their precise distribution between tumor vessels, alveolus and tumor nodules by histological analysis. Finally, we observed that tumor EVs can be used as effective vectors to increase gold nanoparticle delivery towards metastatic nodules. Conclusions Our findings provide a valuable tool to study the distribution and interaction of EVs in mice and a novel strategy to improve the targeting of gold nanoparticles to cancer cells and metastatic nodules by using the natural properties of malignant EVs.
Collapse
Affiliation(s)
- Pablo Lara
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Sujey Palma-Florez
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Edison Salas-Huenuleo
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Iva Polakovicova
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Laboratory of Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago, Chile
| | - Simón Guerrero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Instituto de investigación Interdisciplinar en Ciencias biomédicas, Universidad SEK (I3CBSEK). Facultad Ciencias de La Salud, Fernando Manterola 0789, Santiago, Chile
| | - Lorena Lobos-Gonzalez
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Centro de Medicina Regenerativa, Facultad de Medicina-Clinica Alemana, Universidad Del Desarrollo, Avenida las condes 12438, lo Barnechea, Santiago, Chile
| | - America Campos
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Luis Muñoz
- Laboratorio de Análisis Por Activación Neutrónica, Comisión Chilena de Energía Nuclear, Nueva Bilbao, 12501, Santiago, Chile
| | - Carla Jorquera-Cordero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Manuel Varas-Godoy
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Jorge Cancino
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Eloísa Arias
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Jaime Villegas
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Republica 440, Santiago, Chile
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands
| | - Fernando Albericio
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, and Department of Organic Chemistry, University of Barcelona, 08028, Barcelona, Spain
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Universidad Andres Bello, Republica 275, 8370146, Santiago, Chile
| | - Alejandro H Corvalan
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Laboratory of Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago, Chile
| | - Andrew F G Quest
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.
| | - Marcelo J Kogan
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.
| |
Collapse
|
42
|
Thiam LG, Aniweh Y, Quansah EB, Donkor JK, Gwira TM, Kusi KA, Niang M, Awandare GA. Cell trace far-red is a suitable erythrocyte dye for multi-color Plasmodium falciparum invasion phenotyping assays. Exp Biol Med (Maywood) 2020; 245:11-20. [PMID: 31903776 PMCID: PMC6987746 DOI: 10.1177/1535370219897393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/06/2019] [Indexed: 11/16/2022] Open
Abstract
Plasmodium falciparum erythrocyte invasion phenotyping assays are a very useful tool for assessing parasite diversity and virulence, and for characterizing the formation of ligand–receptor interactions. However, such assays need to be highly sensitive and reproducible, and the selection of labeling dyes for differentiating donor and acceptor erythrocytes is a critical factor. We investigated the suitability of cell trace far-red (CTFR) as a dye for P. falciparum invasion phenotyping assays. Using the dyes carboxyfluorescein diacetate succinimidyl ester (CFDA-SE) and dichloro dimethyl acridin one succinimidyl ester (DDAO-SE) as comparators, we used a dye-dilution approach to assess the limitations and specific staining procedures for the applicability of CTFR in P. falciparum invasion phenotyping assays. Our data show that CTFR effectively labels acceptor erythrocytes and provides a stable fluorescent intensity at relatively low concentrations. CTFR also yielded a higher fluorescence intensity relative to DDAO-SE and with a more stable fluorescence intensity over time. Furthermore, CTFR did not affect merozoites invasion of erythrocytes and was not toxic to the parasite’s intraerythrocytic development. Additionally, CTFR offers flexibility in the choice of combinations with several other DNA dyes, which broaden its usage for P. falciparum erythrocyte invasion assays, considering a wider range of flow cytometers with various laser settings.
Collapse
Affiliation(s)
- Laty G Thiam
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Evelyn B Quansah
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Jacob K Donkor
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Theresa M Gwira
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Kwadwo A Kusi
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
- Department of Immunology, Noguchi Memorial Institute for Medical
Research, University of Ghana, P. O. Box LG 581, Legon, Accra
| | - Makhtar Niang
- Unité d’Immunologie, Institut Pasteur de Dakar, Dakar 220,
Senegal
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| |
Collapse
|
43
|
Yousefi Z, Sharifzadeh S, Yar-Ahmadi V, Andalib A, Eskandari N. Fc Receptor-Like 1 as a Promising Target for Immunotherapeutic Interventions of B-Cell-Related Disorders. Biomark Insights 2019; 14:1177271919882351. [PMID: 31798301 PMCID: PMC6864034 DOI: 10.1177/1177271919882351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/22/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human B-cell responses are regulated through synergy between a collection of activation and inhibitory receptors. Fc receptor-like (FCRL) molecules have recently been identified as co-receptors that are preferentially expressed in human B-cells, which may also play an important role in the regulation of human B-cell responses. FCRL1 is a member of the FCRL family molecules with 2 immunoreceptor tyrosine-based activation motifs (ITAMs) in its cytoplasmic tail. This study aimed to investigate the regulatory roles of FCRL1 in human B-cell responses. MATERIALS AND METHODS The regulatory potential of FCRL1 in human B-cell through knockdown of FCRL1 expression in the Ramos and Daudi Burkitt lymphoma (BL) cell lines by using the retroviral-based short hairpin ribonucleic acid (shRNA) delivery method. The functional consequences of FCRL1 knockdown were assessed by measuring the proliferation, apoptosis, and the expression levels of Bcl-2, Bid, and Bax genes as well as phosphoinositide-3 kinase/-serine-threonine kinase AKT (PI3K/p-AKT) pathway in the BL cells, using the quantitative real-time polymerase chain reaction (PCR) and flow cytometry analysis. The NF-κB activity was also measured by the enzyme-linked immunosorbent assay (ELISA). RESULTS FCRL1 knockdown significantly decreased cell proliferation and increased apoptotic cell death in the BL cells. There was a significant reduction in the extent of the Bcl-2 gene expression in the treated BL cells compared with control cells. On the contrary, FCRL1 knockdown increased the expression levels of Bid and Bax genes in the treated BL cells when compared with control cells. In addition, the extent of the PI3K/p-AKT expression and phosphorylated-p65 NF-κB activity was significantly decreased in the treated BL cells compared with control cells. CONCLUSIONS These results suggest that FCRL1 can play a key role in the activation of human B-cell responses and has the potential to serve as a target for immunotherapy of FCRL1 positive B-cell-related disorders.
Collapse
Affiliation(s)
- Zahra Yousefi
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sedigheh Sharifzadeh
- Diagnostic Laboratory Sciences and
Technology Research Center, School of Paramedical Sciences, Shiraz University of
Medical Sciences, Shiraz, Iran
| | - Vali Yar-Ahmadi
- Department of Parasitology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center,
Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
44
|
Gavin C, Meinke S, Heldring N, Heck KA, Achour A, Iacobaeus E, Höglund P, Le Blanc K, Kadri N. The Complement System Is Essential for the Phagocytosis of Mesenchymal Stromal Cells by Monocytes. Front Immunol 2019; 10:2249. [PMID: 31616424 PMCID: PMC6763726 DOI: 10.3389/fimmu.2019.02249] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/05/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cell (MSC) therapy is a promising tool in the treatment of chronic inflammatory diseases. This has been ascribed to the capacity of MSC to release a large variety of immune-modulatory factors. However, all aspects of the mode of therapeutic MSC action in different diseases remain unresolved, mainly because most of the infused MSC are undetectable in the circulation within hours after infusion. The aim of this study was to elucidate the fate of MSC after contact with plasma. We found that upon contact with blood, complement proteins including C3b/iC3b are deposited on MSC. Importantly, we also found that complement bound to MSC enhanced their phagocytosis by classical and intermediate monocytes via a mechanism that involves C3 but not C5. Thus, we describe for the first time a mechanism which might explain, at least partly, why MSC are not found in the blood circulation after infusion. Our results indicate that MSC immune-modulatory effects could be mediated by monocytes that have phagocytosed them.
Collapse
Affiliation(s)
- Caroline Gavin
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Meinke
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Nina Heldring
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Anne Heck
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ellen Iacobaeus
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Petter Höglund
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.,Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Katarina Le Blanc
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Center of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Nadir Kadri
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Tremp M, Sprenger L, Degrugillier L, Schaefer DJ, Madduri S, Schaeren S, Kalbermatten DF. Regeneration of nerve crush injury using adipose-derived stem cells: A multimodal comparison. Muscle Nerve 2019; 58:566-572. [PMID: 30028517 PMCID: PMC6646906 DOI: 10.1002/mus.26188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/26/2023]
Abstract
Introduction: To restore full function following nerve crush injuries is critical but challenging. In an attempt to develop a viable therapy, we evaluated the effect of rat adipose‐derived stem cells (rASC) in 2 different settings of a sciatic crush injury model. Methods: In the first group, after 14 days of nerve crush injury, rASCs were injected distal to the lesion under ultrasound guidance. In the other group, alleviation of compression through clip removal (CR) was combined with epineural injection of rASCs. Gait analyses, MRI, gastrocnemius muscle weight ratio (MWR), and histomorphometry were performed for outcome analysis. Results: CR combined with rASC injection resulted in less muscle atrophy, as evidenced by MWR. These findings are further supported by better functional and anatomical outcomes. Discussion: Animals treated with CR and epineural stem cell injection showed enhanced anatomical and functional recovery. Muscle Nerve58: 566–572, 2018
Collapse
Affiliation(s)
- Mathias Tremp
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Lima Sprenger
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Lucas Degrugillier
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Dirk Johannes Schaefer
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, Center for Bioengineering and Regenerative Medicine, University of Basel, Gewerbestrasse 14, 4123, Allschwil, Switzerland
| | - Srinivas Madduri
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, Center for Bioengineering and Regenerative Medicine, University of Basel, Gewerbestrasse 14, 4123, Allschwil, Switzerland
| | - Stefan Schaeren
- Department of Spinal Surgery, University Hospital Basel, Basel, Switzerland
| | - Daniel Felix Kalbermatten
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
46
|
Park J, Kwon M, Kim KH, Kim TS, Hong SH, Kim CG, Kang SG, Moon JH, Kim EH, Park SH, Chang JH, Shin EC. Immune Checkpoint Inhibitor-induced Reinvigoration of Tumor-infiltrating CD8 + T Cells is Determined by Their Differentiation Status in Glioblastoma. Clin Cancer Res 2019; 25:2549-2559. [PMID: 30659023 DOI: 10.1158/1078-0432.ccr-18-2564] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/07/2018] [Accepted: 01/16/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune checkpoint inhibitors (ICI) are used for the treatment of various cancers, but clinical trials of anti-programmed cell death protein 1 (PD-1) with patients with recurrent glioblastoma (GBM) have failed to show clinical benefits. In this study, we examined the differentiation status of CD8+ tumor-infiltrating lymphocytes (TIL) from patients with primary GBM and their reinvigoration by ICIs to understand the nature of T-cell exhaustion in GBM. EXPERIMENTAL DESIGN We isolated TILs from 98 patients with newly diagnosed GBM and examined the expression of immune checkpoint receptors and T-cell transcription factors using flow cytometry. TILs were ex vivo stimulated with anti-CD3 in the presence of anti-PD-1 and/or anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) and their proliferation assessed. RESULTS CD8+ TILs had significantly increased expression of immune checkpoint receptors, including PD-1 and CTLA-4, compared with peripheral blood CD8+ T cells. Among CD8+ TILs, PD-1+ cells exhibited more terminally differentiated phenotypes (i.e., EomeshiT-betlo) than PD-1- cells. These data were confirmed by analyzing NY-ESO-1157-specific CD8+ TILs. Evaluating the proliferation of CD8+ TILs after ex vivo stimulation with anti-CD3 and anti-PD-1, we found that proliferation inversely correlated with the percentage of EomeshiT-betlo cells among PD-1+CD8+ TILs. When anti-CTLA-4 was used in combination with anti-PD-1, an additional increase in CD8+ TIL proliferation was observed in patients with low percentages of EomeshiT-betlo CD8+ TILs, who responded well to anti-PD-1 in ex vivo assays, but not in patients with high percentages of EomeshiT-betlo CD8+ TILs, who did not respond to anti-PD-1. CONCLUSIONS In primary GBM, the differentiation status of CD8+ TILs determines their reinvigoration ability upon ICI treatment.
Collapse
Affiliation(s)
- Junsik Park
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Minsuk Kwon
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kyung Hwan Kim
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tae-Shin Kim
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seon-Hui Hong
- BioMedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Chang Gon Kim
- Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hyung Moon
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su-Hyung Park
- Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea. .,BioMedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
47
|
Lachowicz JI, Picci G, Coni P, Lippolis V, Mamusa M, Murgia S, Pichiri G, Caltagirone C. Fluorescent squaramide ligands for cellular imaging and their encapsulation in cubosomes. NEW J CHEM 2019. [DOI: 10.1039/c9nj01548e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two new fluorescent squaramides bearing quinoline (L1) and naphthalene (L2) as fluorogenic fragments were synthesized and investigated as possible cellular imaging probes as free molecules and when loaded in monoolein-based cubosomes.
Collapse
Affiliation(s)
- Joanna I. Lachowicz
- Dipartimento di Scienze Chimiche e Geologiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Giacomo Picci
- Dipartimento di Scienze Chimiche e Geologiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Pierpaolo Coni
- Dipartimento di Scienze Chirurgiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Vito Lippolis
- Dipartimento di Scienze Chimiche e Geologiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Marianna Mamusa
- CSGI
- Department of Chemistry Ugo Shiff
- University of Florence
- I-50019 Sesto Fiorentino
- Italy
| | - Sergio Murgia
- Dipartimento di Scienze Chimiche e Geologiche and CSGI
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Giuseppina Pichiri
- Dipartimento di Scienze Chirurgiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| | - Claudia Caltagirone
- Dipartimento di Scienze Chimiche e Geologiche
- Università degli Studi di Cagliari
- Cittadella Universitaria
- I-09042 Monserrato
- Italy
| |
Collapse
|
48
|
Abstract
CD8+ and CD4+ T cells play a critical role in viral clearance during self-limited HCV infection. Moreover, induction of robust and polyfunctional memory T cells is a primary goal in the development of T-cell-based HCV prophylactic vaccines. Direct ex vivo function of HCV-specific T cells without in vitro expansion can be assessed by measuring cytokine secretion or cell proliferation upon in vitro stimulation with HCV antigens using enzyme-linked immunospot (ELISpot) assays, intracellular cytokine staining (ICS) assays, and flow cytometry-based T-cell proliferation assays. In particular, polyfunctionality of T cells can be assessed by ICS assays using multicolor flow cytometry. Here, we provide protocols to measure direct ex vivo functions of HCV-specific T cells.
Collapse
|
49
|
Chuo STY, Chien JCY, Lai CPK. Imaging extracellular vesicles: current and emerging methods. J Biomed Sci 2018; 25:91. [PMID: 30580764 PMCID: PMC6304785 DOI: 10.1186/s12929-018-0494-5] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanoparticles released by cells. They range from 30 nm to several micrometers in diameter, and ferry biological cargos such as proteins, lipids, RNAs and DNAs for local and distant intercellular communications. EVs have since been found to play a role in development, as well as in diseases including cancers. To elucidate the roles of EVs, researchers have established different methods to visualize and study their spatiotemporal properties. However, since EV are nanometer-sized, imaging them demands a full understanding of each labeling strategy to ensure accurate monitoring. This review covers current and emerging strategies for EV imaging for prospective studies.
Collapse
Affiliation(s)
- Steven Ting-Yu Chuo
- Institute of Atomic and Molecular Sciences, Academia Sinica, No. 1, Roosevelt Rd., Sec. 4, Taipei, 10617 Taiwan
| | - Jasper Che-Yung Chien
- Institute of Atomic and Molecular Sciences, Academia Sinica, No. 1, Roosevelt Rd., Sec. 4, Taipei, 10617 Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, No. 1, Roosevelt Rd., Sec. 4, Taipei, 10617 Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
50
|
TRIM44 promotes quiescent multiple myeloma cell occupancy and survival in the osteoblastic niche via HIF-1α stabilization. Leukemia 2018; 33:469-486. [PMID: 30089913 PMCID: PMC6365383 DOI: 10.1038/s41375-018-0222-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022]
Abstract
Despite progress in the treatment of MM, including the use of high-dose chemotherapy and autologous stem cell transplantation, a considerable proportion of patients are refractory to all therapies. This resistance is related to the molecular genetic heterogeneity in MM cells as well as to the contributions from the BM, which is one of the key determinants of treatment outcome. Our previous studies using fluorescent tracers revealed that MM heterogeneity is correlated with the presence of quiescent stem-like cancer cells, which prefer to reside within the osteoblastic niche of the BM. In this report, we identified a novel protein, tripartite motif containing 44 (TRIM44), which is overexpressed in the osteoblastic niche of the BM, enabling MM cells to compete with HSCs for niche support. TRIM44 expression in MM cells promoted cell quiescence but increased bone destruction in xenograft mice, similar to what is observed in MM patients. TRIM44 functions as a deubiquitinase for hypoxia inducible factor-1α (HIF-1α), which stabilizes HIF-1α expression during hypoxia and normoxia. Stabilized HIF-1α stimulates MM cell growth and survival during hypoxia. Our work is the first report to reveal signaling in quiescent MM cells and the functions of TRIM44.
Collapse
|