51
|
Giuliani G, Merolla A, Paolino M, Reale A, Saletti M, Blancafort L, Cappelli A, Benfenati F, Cesca F. Stability Studies of New Caged bis-deoxy-coelenterazine Derivatives and Their Potential Use as Cellular pH Probes. Photochem Photobiol 2020; 97:343-352. [PMID: 33095933 DOI: 10.1111/php.13347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/16/2020] [Indexed: 11/28/2022]
Abstract
The synthesis of new bis-deoxy-coelenterazine (1) derivatives bearing ester protective groups (acetate, propionate and butyrate esters) was accomplished. Moreover, their hydrolytic stability at room temperature was evaluated in dimethylsulfoxide (DMSO) as solvent, using the nuclear magnetic resonance (NMR) spectra of the key products at different time intervals. The results showed an increasing hydrolysis rate according to longest aliphatic chain, with a half-life of 24 days of the more stable acetate derivative (4a). Furthermore, the analysis of the experimental data revealed the greater stability of the enol tautomer in this aprotic polar solvent. This result was confirmed by theoretical calculations using the density functional theory (DFT) approach, which gave us the opportunity to propose a detailed decomposition mechanism. Additionally, the derivatives obtained were tested by bioluminescence luciferase assays to evaluate their potential use as extracellular pH-sensitive reporter substrates of luciferase. The biological data support the idea that further structural modifications of these molecules may open promising perspectives in this field of research.
Collapse
Affiliation(s)
- Germano Giuliani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Assunta Merolla
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Marco Paolino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Annalisa Reale
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Mario Saletti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lluís Blancafort
- Departament de Química, Facultat de Ciències, Institut de Química Computacional i Catàlisi (IQCC), Universitat de Girona, Girona, Spain
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
52
|
Mizui Y, Eguchi M, Tanaka M, Ikeda Y, Yoshimura H, Ozawa T, Citterio D, Hiruta Y. Long-term single cell bioluminescence imaging with C-3 position protected coelenterazine analogues. Org Biomol Chem 2020; 19:579-586. [PMID: 33140803 DOI: 10.1039/d0ob02020f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bioluminescence is a powerful imaging modality for monitoring biological phenomena both in vitro and in vivo. Bioluminescence imagin (BLI) is becoming a seamless imaging technology covering the range from cells to organs of small animals. Long-term imaging at the single cell level would lead to a true understanding of the dynamics of life phenomena. This work presents a long-term single cell bioluminescence imaging technology accomplished with C-3 position protected furimazines (FMZs), a CTZ analogues, which generate intense blue emission when paired with a highly stable engineered luciferase, Nanoluc. Four types of FMZs protected at the C-3 position have been synthesized. The type and steric bulkiness of the protection group strongly contributed to storage stability and the kinetics of the bioluminescence reactions of the analogues in human living cells. In particular, two developed FMZ analogues resulted in significantly longer bioluminescence emission with higher S/N ratio than FMZ at single cell level. Long-term bioluminescence single cell imaging technology with the developed FMZ analogues will lead to seamless imaging in the range from cells to organs of small animals.
Collapse
Affiliation(s)
- Yuki Mizui
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Dimond A, Van de Pette M, Fisher AG. Illuminating Epigenetics and Inheritance in the Immune System with Bioluminescence. Trends Immunol 2020; 41:994-1005. [PMID: 33036908 DOI: 10.1016/j.it.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
The remarkable process of light emission by living organisms has fascinated mankind for thousands of years. A recent expansion in the repertoire of catalytic luciferase enzymes, coupled with the discovery of the genes and pathways that encode different luciferin substrates, means that bioluminescence imaging (BLI) is set to revolutionize longitudinal and dynamic studies of gene control within biomedicine, including the regulation of immune responses. In this review article, we summarize recent advances in bioluminescence-based imaging approaches that promise to enlighten our understanding of in vivo gene and epigenetic control within the immune system.
Collapse
Affiliation(s)
- Andrew Dimond
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mathew Van de Pette
- Epigenetic Mechanisms of Toxicity, MRC Toxicology Unit, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Amanda G Fisher
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
54
|
Zhang W, Zhang X, Zhang Y, Zhang X, Zou T, Zhao W, Lv Y, Wang J, Dai P, Cui H, Zhang Y, Gao D, Ruan C, Zhang X. Retracted: Cell Fate and Tissue Remodeling in Canine Urethral Repair Using a Bone Marrow Mesenchymal Stem Cell+Endothelial Progenitor Cell Amniotic Patch. Tissue Eng Part A 2020; 26:e1403-e1412. [PMID: 32808578 DOI: 10.1089/ten.tea.2020.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The Editors of Tissue Engineering: Part A retract the article entitled, "Cell Fate and Tissue Remodeling in Canine Urethral Repair Using a Bone Marrow Mesenchymal Stem Cell+Endothelial Progenitor Cell Amniotic Patch," by Wenxin Zhang, Xin Zhang, Yihua Zhang, Xinke Zhang, Tong Zou, Wen Zhao, Yangou Lv, Jinglu Wang, Pengxiu Dai, Hao Cui, Yi Zhang, Dengke Gao, Chenmei Ruan, and Xia Zhang (epub ahead of print September 21, 2020; DOI: http://doi.org/10.1089/ten.tea.2020.0129). After the online publication of the article, the authors have indicated that they "feel that we have not yet studied our work completely and some new great results are discovered. So after carefully thinking, we are going to rearrange this manuscript and try to give more precise model. [sic]" The authors have not explained what those expected results will be, so it remains unclear the direction their work is headed. The authors also indicated that they plan to submit an updated version of the paper to Tissue Engineering in the future. Upon submission the new manuscript will undergo rigorous peer review, and there is no guarantee of acceptance.
Collapse
Affiliation(s)
- Wenxin Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Xin Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Yihua Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Xinke Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Tong Zou
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Wen Zhao
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Yangou Lv
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Jinglu Wang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Pengxiu Dai
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Hao Cui
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Yi Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Dengke Gao
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Chenmei Ruan
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| | - Xia Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Shaanxi, China
| |
Collapse
|
55
|
Nannan L, Oudart JB, Monboisse JC, Ramont L, Brassart-Pasco S, Brassart B. Extracellular Vesicle-Dependent Cross-Talk in Cancer-Focus on Pancreatic Cancer. Front Oncol 2020; 10:1456. [PMID: 32974169 PMCID: PMC7466446 DOI: 10.3389/fonc.2020.01456] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/09/2020] [Indexed: 01/10/2023] Open
Abstract
Extracellular vesicles (EVs) like exosomes and shed microvesicles are generated by many different cells. However, among all the cells, cancer cells are now recognized to secrete more EVs than healthy cells. Tumor-derived EVs can be isolated from biofluids such as blood, urine, ascitic fluid, and saliva. Their numerous components (nucleic acids, proteins, and lipids) possess many pleiotropic functions involved in cancer progression. The tumor-derived EVs generated under the influence of tumor microenvironment play distant roles and promote cellular communication by directly interacting with different cells. Moreover, they modulate extracellular matrix remodeling and tumor progression. Tumor-derived EVs are involved in pre-metastatic niche formation, dependent on the EV-associated protein receptors, and in cancer chemoresistance as they transfer drug-resistance-related genes to recipient cells. Recent advances in preclinical and clinical fields suggest their potential use as biomarkers for diagnosis and prognosis as well as for drug delivery in cancer. In this Review, we discuss EV characteristics and pro-tumor capacities, and highlight the future crucial impact of tumor-derived EVs in pancreatic cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Lise Nannan
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,Biomedical MRI Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jean-Baptiste Oudart
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Jean Claude Monboisse
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Laurent Ramont
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Sylvie Brassart-Pasco
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
56
|
Li H, Yuan L, Long Y, Fang H, Li M, Liu Q, Xia X, Qin C, Zhang Y, Lan X, Gai Y. Synthesis and Preclinical Evaluation of a 68Ga-Radiolabeled Peptide Targeting Very Late Antigen-3 for PET Imaging of Pancreatic Cancer. Mol Pharm 2020; 17:3000-3008. [PMID: 32544337 DOI: 10.1021/acs.molpharmaceut.0c00416] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pancreatic cancer is highly malignant and has a five-year survival rate of 5% due to an early lymph node, nerve, and vascular metastasis. Integrin α3β1 (also called very late antigen-3, VLA-3) is overexpressed in many tumors and plays a vital role in tumor formation, recurrence, and metastasis. In this study, we developed a 68Ga-radiolabeled peptide tracer targeting the α3 unit of VLA-3 and evaluated its potential application in positron emission computed tomography (PET) imaging of pancreatic cancer. NOTA-CK11 was prepared by solid-phase synthesis and successfully radiolabeled with 68Ga with greater than 99% radiochemical purity and a specific activity of 37 ± 5 MBq/nmol (n = 5). The expression level of integrin α3 in three human pancreatic cancer cells was evaluated with the order of SW1990, BXPC-3, and PANC-1 from high to low, while the expression level of integrin β1 was relatively close. When SW1990 cells with the highest expression level of VLA-3 were stained with FITC-CK11, strong fluorescence was observed by flow cytometry and under a laser confocal microscope. However, no significant fluorescence was observed in the blocking group when treated with excessive CK11. 68Ga-NOTA-CK11 showed significant radioactivity accumulation in SW1990 cells and was blocked by CK11 successfully. Subsequent small-animal PET imaging and biodistribution studies in mice bearing SW1990 xenografts confirmed its high tumor uptake with a good tumor-to-blood ratio and tumor-to-muscle ratio (2.45 ± 0.31 and 3.65 ± 0.33, respectively) at 1 h post injection of the probe. In summary, we successfully developed a peptide-based imaging agent, 68Ga-NOTA-CK11, that showed a strong binding affinity with VLA-3 and good target specificity for SW1990 cells and xenografted pancreatic tumor, rending it a promising radiotracer for PET imaging of VLA-3 expression in pancreatic cancer.
Collapse
Affiliation(s)
- Huiling Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lujie Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
57
|
Mastraccio KE, Huaman C, Warrilow D, Smith GA, Craig SB, Weir DL, Laing ED, Smith IL, Broder CC, Schaefer BC. Establishment of a longitudinal pre-clinical model of lyssavirus infection. J Virol Methods 2020; 281:113882. [PMID: 32407866 PMCID: PMC8056983 DOI: 10.1016/j.jviromet.2020.113882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/19/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Traditional mouse models of lyssavirus pathogenesis rely on euthanizing large groups of animals at various time points post-infection, processing infected tissues, and performing histological and molecular analyses to determine anatomical sites of infection. While powerful by some measures, this approach is limited by the inability to monitor disease progression in the same mice over time. In this study, we established a novel non-invasive mouse model of lyssavirus pathogenesis, which consists of longitudinal imaging of a luciferase-expressing Australian bat lyssavirus (ABLV) reporter virus. In vivo bioluminescence imaging (BLI) in mice revealed viral spread from a peripheral site of inoculation into the central nervous system (CNS), with kinetically and spatially distinct foci of replication in the footpad, spinal cord, and hindbrain. Detection of virus within the CNS was associated with onset of clinical disease. Quantification of virus-derived luminescent signal in the brain was found to be a reliable measure of viral replication, when compared to traditional molecular methods. Furthermore, we demonstrate that in vivo imaging of ABLV infection is not restricted to the use of albino strains of mice, but rather strong BLI signal output can be achieved by shaving the hair from the heads and spines of pigmented strains, such as C57BL/6. Overall, our data show that in vivo BLI can be used to rapidly and non-invasively identify sites of lyssavirus replication and to semi-quantitatively determine viral load without the need to sacrifice mice at multiple time points.
Collapse
Affiliation(s)
- Kate E Mastraccio
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Celeste Huaman
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - David Warrilow
- Queensland Health Forensic and Scientific Services, Archerfield, Australia.
| | - Greg A Smith
- Queensland Health Forensic and Scientific Services, Archerfield, Australia.
| | - Scott B Craig
- Queensland Health Forensic and Scientific Services, Archerfield, Australia.
| | - Dawn L Weir
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Eric D Laing
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Ina L Smith
- Queensland Health Forensic and Scientific Services, Archerfield, Australia; Risk Evaluation and Preparedness Program, Health and Biosecurity, CSIRO, Black Mountain, ACT, Australia.
| | - Christopher C Broder
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Brian C Schaefer
- Uniformed Services University, Department of Microbiology and Immunology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
58
|
Shin N, Lee SH, Pham Ba VA, Park TH, Hong S. Micelle-stabilized Olfactory Receptors for a Bioelectronic Nose Detecting Butter Flavors in Real Fermented Alcoholic Beverages. Sci Rep 2020; 10:9064. [PMID: 32493940 PMCID: PMC7270175 DOI: 10.1038/s41598-020-65900-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/05/2020] [Indexed: 11/09/2022] Open
Abstract
A bioelectronic nose device based on micelle-stabilized olfactory receptors is developed for the selective discrimination of a butter flavor substance in commercial fermented alcoholic beverages. In this work, we have successfully overexpressed ODR-10, a type of olfactory receptor, from Caenorhabditis elegans using a bacterial expression system at a low cost and high productivity. The highly-purified ODR-10 was stabilized in micelle structures, and it was immobilized on a carbon nanotube field-effect transistor to build a bioelectronic nose for the detection of diacetyl, a butter flavor substance, via the specific interaction between diacetyl and ODR-10. The bioelectronic nose device can sensitively detect diacetyl down to 10 fM, and selectively discriminate it from other substances. In addition, this sensor could directly evaluate diacetyl levels in a variety of real fermented alcoholic beverages such as beer, wine, and makgeolli (fermented Korean wine), while the sensor did not respond to soju (Korean style liquor without diacetyl). In this respect, our sensor should be a powerful tool for versatile food industrial applications such as the quality control of alcoholic beverages and foods.
Collapse
Affiliation(s)
- Narae Shin
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea
| | - Seung Hwan Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Korea.,Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15588, Korea
| | - Viet Anh Pham Ba
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea.,Department of Environmental Toxicology and Monitoring, Hanoi University of Natural Resources and Environment, Hanoi, Vietnam
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Korea.
| | - Seunghun Hong
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
59
|
Gangadaran P, Rajendran RL, Ahn BC. Application of In Vivo Imaging Techniques for Monitoring Natural Killer Cell Migration and Tumor Infiltration. Cancers (Basel) 2020; 12:1318. [PMID: 32455886 PMCID: PMC7281416 DOI: 10.3390/cancers12051318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022] Open
Abstract
In recent years, the use of natural killer (NK) cell-based immunotherapy has shown promise against various cancer types. To some extent therapeutic potential of NK cell-based immunotherapy depends on migration of NK cells towards tumors in animal models or human subjects and subsequent infiltration. Constant improvement in the pharmacological and therapeutic properties of NK cells is driving the performance and use of NK cell-based immunotherapies. In this review, we summarize the molecular imaging techniques used in monitoring the migration and infiltration of NK cells in vivo at preclinical and clinical levels. A review of pros and cons of each molecular imaging modality is done. Finally, we provide our perception of the usefulness of molecular imaging approaches for in vivo monitoring of NK cells in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (R.L.R.)
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (R.L.R.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (R.L.R.)
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| |
Collapse
|
60
|
Pancreatic Cancer UK Grand Challenge: Developments and challenges for effective CAR T cell therapy for pancreatic ductal adenocarcinoma. Pancreatology 2020; 20:394-408. [PMID: 32173257 DOI: 10.1016/j.pan.2020.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Death from pancreatic ductal adenocarcinoma (PDAC) is rising across the world and PDAC is predicted to be the second most common cause of cancer death in the USA by 2030. Development of effective biotherapies for PDAC are hampered by late presentation, a low number of differentially expressed molecular targets and a tumor-promoting microenvironment that forms both a physical, collagen-rich barrier and is also immunosuppressive. In 2017 Pancreatic Cancer UK awarded its first Grand Challenge Programme award to tackle this problem. The team plan to combine the use of novel CAR T cells with strategies to overcome the barriers presented by the tumor microenvironment. In advance of publication of those data this review seeks to highlight the key problems in effective CAR T cell therapy of PDAC and to describe pre-clinical and clinical progress in CAR T bio-therapeutics.
Collapse
|
61
|
Chen G, Cao Y, Tang Y, Yang X, Liu Y, Huang D, Zhang Y, Li C, Wang Q. Advanced Near-Infrared Light for Monitoring and Modulating the Spatiotemporal Dynamics of Cell Functions in Living Systems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903783. [PMID: 32328436 PMCID: PMC7175256 DOI: 10.1002/advs.201903783] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Indexed: 05/07/2023]
Abstract
Light-based technique, including optical imaging and photoregulation, has become one of the most important tools for both fundamental research and clinical practice, such as cell signal sensing, cancer diagnosis, tissue engineering, drug delivery, visual regulation, neuromodulation, and disease treatment. In particular, low energy near-infrared (NIR, 700-1700 nm) light possesses lower phototoxicity and higher tissue penetration depth in living systems as compared with ultraviolet/visible light, making it a promising tool for in vivo applications. Currently, the NIR light-based imaging and photoregulation strategies have offered a possibility to real-time sense and/or modulate specific cellular events in deep tissues with subcellular accuracy. Herein, the recent progress with respect to NIR light for monitoring and modulating the spatiotemporal dynamics of cell functions in living systems are summarized. In particular, the applications of NIR light-based techniques in cancer theranostics, regenerative medicine, and neuroscience research are systematically introduced and discussed. In addition, the challenges and prospects for NIR light-based cell sensing and regulating techniques are comprehensively discussed.
Collapse
Affiliation(s)
- Guangcun Chen
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yuheng Cao
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Yanxing Tang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Xue Yang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yongyang Liu
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Dehua Huang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yejun Zhang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Chunyan Li
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
- College of Materials Sciences and Opto‐Electronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
62
|
Wong RM, Li TK, Li J, Ho WT, Chow SKH, Leung SS, Cheung WH, Ip M. A systematic review on current osteosynthesis-associated infection animal fracture models. J Orthop Translat 2020; 23:8-20. [PMID: 32440511 PMCID: PMC7231979 DOI: 10.1016/j.jot.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Osteosynthesis-associated infection is a challenging complication post fracture fixation, burdening the patients and the orthopaedic surgeons alike. A clinically relevant animal model is critical in devising new therapeutic strategies. Our aim was to perform a systematic review to evaluate existing preclinical models and identify their applications in aspects of animal selection, bacterial induction, fracture fixation and complications. Methods A systematic literature research was conducted in PubMed and Embase up to February 2020. A total of 31 studies were included. Information on the animal, bacterial induction, fracture fixation, healing result and complications were extracted. Results Animals selected included murine (23), rabbit (6), ewe (1) and goat (1). Larger animals had enabled the use of human-sized implant, however small animals were more economical and easier in handling. Staphylococcus aureus (S. aureus) was the most frequently chosen bacteria for induction. Bacterial inoculation dose ranged from 102-8 CFU. Consistent and replicable infections were observed from 104 CFU in general. Methods of inoculation included injections of bacterial suspension (20), placement of foreign objects (8) and pretreatment of implants with established biofilm (3). Intramedullary implants (13), plates and screws (18) were used in most models. Radiological (29) and histological evaluations (24) in osseous healing were performed. Complications such as instability of fracture fixation (7), unexpected surgical death (5), sepsis (1) and persistent lameness (1) were encountered. Conclusion The most common animal model is the S. aureus infected open fracture internally fixated. Replicable infections were mainly from 104 CFU of bacteria. However, with the increase in antibiotic resistance, future directions should explore polymicrobial and antibiotic resistant strains, as these will no doubt play a major role in bone infection. Currently, there is also a lack of osteoporotic bone infection models and the pathophysiology is unexplored, which would be important with our aging population. The translational potential of this article This systematic review provides an updated overview and compares the currently available animal models of osteosynthesis-associated infections. A discussion on future research directions and suggestion of animal model settings were made, which is expected to advance the research in this field.
Collapse
Affiliation(s)
- Ronald M.Y. Wong
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Tsz-kiu Li
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Jie Li
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Wing-Tung Ho
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Simon K.-H. Chow
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | | | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
- Corresponding author. Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Margaret Ip
- Department of Microbiology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
63
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
64
|
Le D, Dhamecha D, Gonsalves A, Menon JU. Ultrasound-Enhanced Chemiluminescence for Bioimaging. Front Bioeng Biotechnol 2020; 8:25. [PMID: 32117914 PMCID: PMC7016203 DOI: 10.3389/fbioe.2020.00025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Tissue imaging has emerged as an important aspect of theragnosis. It is essential not only to evaluate the degree of the disease and thus provide appropriate treatments, but also to monitor the delivery of administered drugs and the subsequent recovery of target tissues. Several techniques including magnetic resonance imaging (MRI), computational tomography (CT), acoustic tomography (AT), biofluorescence (BF) and chemiluminescence (CL), have been developed to reconstruct three-dimensional images of tissues. While imaging has been achieved with adequate spatial resolution for shallow depths, challenges still remain for imaging deep tissues. Energy loss is usually observed when using a magnetic field or traditional ultrasound (US), which leads to a need for more powerful energy input. This may subsequently result in tissue damage. CT requires exposure to radiation and a high dose of contrast agent to be administered for imaging. The BF technique, meanwhile, is affected by strong scattering of light and autofluorescence of tissues. The CL is a more selective and sensitive method as stable luminophores are produced from physiochemical reactions, e.g. with reactive oxygen species. Development of near infrared-emitting luminophores also bring potential for application of CL in deep tissues and whole animal studies. However, traditional CL imaging requires an enhancer to increase the intensity of low-level light emissions, while reducing the scattering of emitted light through turbid tissue environment. There has been interest in the use of focused ultrasound (FUS), which can allow acoustic waves to propagate within tissues and modulate chemiluminescence signals. While light scattering is decreased, the spatial resolution is increased with the assistance of US. In this review, chemiluminescence detection in deep tissues with assistance of FUS will be highlighted to discuss its potential in deep tissue imaging.
Collapse
Affiliation(s)
| | | | | | - Jyothi U. Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
65
|
Application of molecular imaging technology in tumor immunotherapy. Cell Immunol 2020; 348:104039. [DOI: 10.1016/j.cellimm.2020.104039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/21/2019] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
|
66
|
Moussa Z, Judeh ZMA, El‐Sharief MAMS, El‐Sharief AMS. N
‐Arylcyanothioformamides: Preparation Methods and Application in the Synthesis of Bioactive Molecules. ChemistrySelect 2020. [DOI: 10.1002/slct.201903534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ziad Moussa
- Department of Chemistry College of Science United Arab Emirates University P.O. Box 15551 Al Ain United Arab Emirates
| | - Zaher M. A. Judeh
- School of Chemical and Biomedical Engineering Nanyang Technological University 62 Nanyang Drive, N1.2-B1-14 Singapore
| | - Marwa A. M. Sh. El‐Sharief
- Applied Organic Chemistry Department National Research Centre, Cairo Egypt
- Faculty of Science and Arts Mohail Asser, King Khalid University Saudi Arabia
| | | |
Collapse
|
67
|
Ni JS, Li Y, Yue W, Liu B, Li K. Nanoparticle-based Cell Trackers for Biomedical Applications. Theranostics 2020; 10:1923-1947. [PMID: 32042345 PMCID: PMC6993224 DOI: 10.7150/thno.39915] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
The continuous or real-time tracking of biological processes using biocompatible contrast agents over a certain period of time is vital for precise diagnosis and treatment, such as monitoring tissue regeneration after stem cell transplantation, understanding the genesis, development, invasion and metastasis of cancer and so on. The rationally designed nanoparticles, including aggregation-induced emission (AIE) dots, inorganic quantum dots (QDs), nanodiamonds, superparamagnetic iron oxide nanoparticles (SPIONs), and semiconducting polymer nanoparticles (SPNs), have been explored to meet this urgent need. In this review, the development and application of these nanoparticle-based cell trackers for a variety of imaging technologies, including fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, magnetic particle imaging, positron emission tomography and single photon emission computing tomography are discussed in detail. Moreover, the further therapeutic treatments using multi-functional trackers endowed with photodynamic and photothermal modalities are also introduced to provide a comprehensive perspective in this promising research field.
Collapse
Affiliation(s)
- Jen-Shyang Ni
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Yaxi Li
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Wentong Yue
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore
| | - Kai Li
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
68
|
Crowley C, Butler CR, Camilli C, Hynds RE, Kolluri KK, Janes SM, De Coppi P, Urbani L. Non-Invasive Longitudinal Bioluminescence Imaging of Human Mesoangioblasts in Bioengineered Esophagi. Tissue Eng Part C Methods 2020; 25:103-113. [PMID: 30648471 PMCID: PMC6389770 DOI: 10.1089/ten.tec.2018.0351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Esophageal engineering aims to create replacement solutions by generating hollow organs using a combination of cells, scaffolds, and regeneration-stimulating factors. Currently, the fate of cells on tissue-engineered grafts is generally determined retrospectively by histological analyses. Unfortunately, quality-controlled cell seeding protocols for application in human patients are not standard practice. As such, the field requires simple, fast, and reliable techniques for non-invasive, highly specific cell tracking. Here, we show that bioluminescence imaging (BLI) is a suitable method to track human mesoangioblast seeding of an esophageal tubular construct at every stage of the preclinical bioengineering pipeline. In particular, validation of BLI as longitudinal quantitative assessment of cell density, proliferation, seeding efficiency, bioreactor culture, and cell survival upon implantation in vivo was performed against standard methods in 2D cultures and in 3D decellularized esophageal scaffolds. The technique is simple, non-invasive, and provides information on mesoangioblast distribution over entire scaffolds. Bioluminescence is an invaluable tool in the development of complex bioartificial organs and can assist in the development of standardized cell seeding protocols, with the ability to track cells from bioreactor through to implantation.
Collapse
Affiliation(s)
- Claire Crowley
- 1 Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health and Great Ormond Street Children's Hospital, University College London, London, United Kingdom
| | - Colin R Butler
- 1 Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health and Great Ormond Street Children's Hospital, University College London, London, United Kingdom.,2 Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Carlotta Camilli
- 1 Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health and Great Ormond Street Children's Hospital, University College London, London, United Kingdom
| | - Robert E Hynds
- 2 Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Krishna K Kolluri
- 2 Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Sam M Janes
- 2 Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Paolo De Coppi
- 1 Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health and Great Ormond Street Children's Hospital, University College London, London, United Kingdom
| | - Luca Urbani
- 1 Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health and Great Ormond Street Children's Hospital, University College London, London, United Kingdom.,3 Institute of Hepatology London, Foundation for Liver Research, London, United Kingdom
| |
Collapse
|
69
|
Tunç E. Biyolüminesans ışıma ve biyolüminesans görüntüleme tekniklerinin moleküler biyoloji araştırmaları bakımından önemi. CUKUROVA MEDICAL JOURNAL 2019. [DOI: 10.17826/cumj.535811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
70
|
Betzer O, Barnoy E, Sadan T, Elbaz I, Braverman C, Liu Z, Popovtzer R. Advances in imaging strategies for in vivo tracking of exosomes. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1594. [PMID: 31840427 DOI: 10.1002/wnan.1594] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
Exosomes have many biological functions as short- and long distance nanocarriers for cell-to-cell communication. They allow the exchange of complex information between cells, and thereby modulate various processes such as homeostasis, immune response and angiogenesis, in both physiological and pathological conditions. In addition, due to their unique abilities of migration, targeting, and selective internalization into specific cells, they are promising delivery vectors. As such, they provide a potentially new field in diagnostics and treatment, and may serve as an alternative to cell-based therapeutic approaches. However, a major drawback for translating exosome treatment to the clinic is that current understanding of these endogenous vesicles is insufficient, especially in regards to their in vivo behavior. Tracking exosomes in vivo can provide important knowledge regarding their biodistribution, migration abilities, toxicity, biological role, communication capabilities, and mechanism of action. Therefore, the development of efficient, sensitive and biocompatible exosome labeling and imaging techniques is highly desired. Recent studies have developed different methods for exosome labeling and imaging, which have allowed for in vivo investigation of their bio-distribution, physiological functions, migration, and targeting mechanisms. These improved imaging capabilities are expected to greatly advance exosome-based nanomedicine applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Oshra Betzer
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel.,Institute of Functional Nano and Soft Materials (FUNSOM), College of Nano Science and Technology (CNST), Soochow University, Suzhou, China
| | - Eran Barnoy
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel
| | - Tamar Sadan
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel
| | - Idan Elbaz
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel
| | - Cara Braverman
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), College of Nano Science and Technology (CNST), Soochow University, Suzhou, China
| | - Rachela Popovtzer
- Faculty of Engineering, Institute of Nanotechnology and Advanced Materials Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
71
|
Kallmeyer K, André-Lévigne D, Baquié M, Krause KH, Pepper MS, Pittet-Cuénod B, Modarressi A. Fate of systemically and locally administered adipose-derived mesenchymal stromal cells and their effect on wound healing. Stem Cells Transl Med 2019; 9:131-144. [PMID: 31613054 PMCID: PMC6954716 DOI: 10.1002/sctm.19-0091] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
There is increasing interest in the use of adipose‐derived mesenchymal stromal cells (ASCs) for wound repair. As the fate of administered cells is still poorly defined, we aimed to establish the location, survival, and effect of ASCs when administered either systemically or locally during wound repair under physiological conditions. To determine the behavior of ASCs, a rat model with wounds on the dorsal aspect of the hind paws was used and two treatment modes were assessed: ASCs administered systemically into the tail vein or locally around the wound. ASCs were transduced to express both firefly luciferase (Fluc) and green fluorescent protein to enable tracking by bioluminescence imaging and immunohistological analysis. Systemically administered ASCs were detected in the lungs 3 hours after injection with a decrease in luminescent signal at 48 hours and signal disappearance from 72 hours. No ASCs were detected in the wound. Locally administered ASCs remained strongly detectable for 7 days at the injection site and became distributed within the wound bed as early as 24 hours post injection with a significant increase observed at 72 hours. Systemically administered ASCs were filtered out in the lungs, whereas ASCs administered locally remained and survived not only at the injection site but were also detected within the wound bed. Both treatments led to enhanced wound closure. It appears that systemically administered ASCs have the potential to enhance wound repair distally from their site of entrapment in the lungs whereas locally administered ASCs enhanced wound repair as they became redistributed within the wound bed.
Collapse
Affiliation(s)
- Karlien Kallmeyer
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland.,Institute for Cellular and Molecular Medicine (ICMM), Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, South Africa
| | - Dominik André-Lévigne
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| | | | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine (ICMM), Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, South Africa.,Department of Human Genetics and Development, University of Geneva, Geneva, Switzerland
| | - Brigitte Pittet-Cuénod
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| | - Ali Modarressi
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| |
Collapse
|
72
|
Fath-Bayati L, Vasei M, Sharif-Paghaleh E. Optical fluorescence imaging with shortwave infrared light emitter nanomaterials for in vivo cell tracking in regenerative medicine. J Cell Mol Med 2019; 23:7905-7918. [PMID: 31559692 PMCID: PMC6850965 DOI: 10.1111/jcmm.14670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/13/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
In vivo tracking and monitoring of adoptive cell transfer has a distinct importance in cell‐based therapy. There are many imaging modalities for in vivo monitoring of biodistribution, viability and effectiveness of transferred cells. Some of these procedures are not applicable in the human body because of low sensitivity and high possibility of tissue damages. Shortwave infrared region (SWIR) imaging is a relatively new technique by which deep biological tissues can be potentially visualized with high resolution at cellular level. Indeed, scanning of the electromagnetic spectrum (beyond 1000 nm) of SWIR has a great potential to increase sensitivity and resolution of in vivo imaging for various human tissues. In this review, molecular imaging modalities used for monitoring of biodistribution and fate of administered cells with focusing on the application of non‐invasive optical imaging at shortwave infrared region are discussed in detail.
Collapse
Affiliation(s)
- Leyla Fath-Bayati
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Tissue Engineering, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Vasei
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Cell-based Therapies Research Institute, Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Imaging Chemistry and Biology, Faculty of Life Sciences and Medicine, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
73
|
Su LJ, Lin HH, Wu MS, Pan L, Yadav K, Hsu HH, Ling TY, Chen YT, Chang HC. Intracellular Delivery of Luciferase with Fluorescent Nanodiamonds for Dual-Modality Imaging of Human Stem Cells. Bioconjug Chem 2019; 30:2228-2237. [PMID: 31268690 DOI: 10.1021/acs.bioconjchem.9b00458] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Delivering functional proteins (such as enzymes) into cells is important in various biological studies and is often accomplished indirectly by transfection with DNA or mRNA encoding recombinant proteins. However, the transfection efficiency of conventional plasmid methods is low for primary cells, which are crucial sources of cell therapy. Here, we present a new platform based on the use of fluorescent nanodiamond (FND) as a biocompatible nanocarrier to enable rapid, effective, and homogeneous labeling of human mesenchymal stem cells (MSCs) with luciferase for multiplex assays and ultrasensitive detection. More than 100 pg of FND and 100 million copies of firefly luciferase can be delivered into each MSC through endocytosis. Moreover, these endocytic luciferase molecules are catalytically active for hours, allowing the cells to be imaged and tracked in vitro as well as in vivo by both fluorescence and bioluminescence imaging. Our results demonstrate that luciferase-conjugated FNDs are useful as multifunctional labels of human stem cells for diverse theranostic applications.
Collapse
Affiliation(s)
- Long-Jyun Su
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan.,Department of Chemistry , National Taiwan University , Taipei 106 , Taiwan
| | - Hsin-Hung Lin
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan
| | - Meng-Shiue Wu
- Department of Pharmacology , National Taiwan University , Taipei 100 , Taiwan
| | - Lei Pan
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan
| | - Kanchan Yadav
- Department of Chemistry , National Taiwan University , Taipei 106 , Taiwan
| | - Hsao-Hsun Hsu
- Department of Surgery, College of Medicine and the Hospital , National Taiwan University , Taipei 100 , Taiwan
| | - Thai-Yen Ling
- Department of Pharmacology , National Taiwan University , Taipei 100 , Taiwan
| | - Yit-Tsong Chen
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan.,Department of Chemistry , National Taiwan University , Taipei 106 , Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan.,Department of Chemical Engineering , National Taiwan University of Science and Technology , Taipei 106 , Taiwan
| |
Collapse
|
74
|
Gil CJ, Tomov ML, Theus AS, Cetnar A, Mahmoudi M, Serpooshan V. In Vivo Tracking of Tissue Engineered Constructs. MICROMACHINES 2019; 10:E474. [PMID: 31315207 PMCID: PMC6680880 DOI: 10.3390/mi10070474] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/10/2019] [Accepted: 07/13/2019] [Indexed: 02/06/2023]
Abstract
To date, the fields of biomaterials science and tissue engineering have shown great promise in creating bioartificial tissues and organs for use in a variety of regenerative medicine applications. With the emergence of new technologies such as additive biomanufacturing and 3D bioprinting, increasingly complex tissue constructs are being fabricated to fulfill the desired patient-specific requirements. Fundamental to the further advancement of this field is the design and development of imaging modalities that can enable visualization of the bioengineered constructs following implantation, at adequate spatial and temporal resolution and high penetration depths. These in vivo tracking techniques should introduce minimum toxicity, disruption, and destruction to treated tissues, while generating clinically relevant signal-to-noise ratios. This article reviews the imaging techniques that are currently being adopted in both research and clinical studies to track tissue engineering scaffolds in vivo, with special attention to 3D bioprinted tissue constructs.
Collapse
Affiliation(s)
- Carmen J Gil
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Martin L Tomov
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Andrea S Theus
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Alexander Cetnar
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30309, USA.
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
75
|
Gálisová A, Herynek V, Swider E, Sticová E, Pátiková A, Kosinová L, Kříž J, Hájek M, Srinivas M, Jirák D. A Trimodal Imaging Platform for Tracking Viable Transplanted Pancreatic Islets In Vivo: F-19 MR, Fluorescence, and Bioluminescence Imaging. Mol Imaging Biol 2019; 21:454-464. [PMID: 30167995 PMCID: PMC6525139 DOI: 10.1007/s11307-018-1270-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Combining specific and quantitative F-19 magnetic resonance imaging (MRI) with sensitive and convenient optical imaging provides complementary information about the distribution and viability of transplanted pancreatic islet grafts. In this study, pancreatic islets (PIs) were labeled with positively charged multimodal nanoparticles based on poly(lactic-co-glycolic acid) (PLGA-NPs) with encapsulated perfluoro-15-crown-5-ether and the near-infrared fluorescent dye indocyanine green. PROCEDURES One thousand and three thousand bioluminescent PIs were transplanted into subcutaneous artificial scaffolds, which served as an alternative transplant site. The grafts were monitored using in vivo F-19 MR, fluorescence, and bioluminescence imaging in healthy rats for 2 weeks. RESULTS Transplanted PIs were unambiguously localized in the scaffolds by F-19 MRI throughout the whole experiment. Fluorescence was detected in the first 4 days after transplantation only. Importantly, in vivo bioluminescence correlated with the F-19 MRI signal. CONCLUSIONS We developed a trimodal imaging platform for in vivo examination of transplanted PIs. Fluorescence imaging revealed instability of the fluorescent dye and its limited applicability for longitudinal in vivo studies. A correlation between the bioluminescence signal and the F-19 MRI signal indicated the fast clearance of PLGA-NPs from the transplantation site after cell death, which addresses a major issue with intracellular imaging labels. Therefore, the proposed PLGA-NP platform is reliable for reflecting the status of transplanted PIs in vivo.
Collapse
Affiliation(s)
- A Gálisová
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - V Herynek
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - E Swider
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - E Sticová
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Pathology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - A Pátiková
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - L Kosinová
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - J Kříž
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - M Hájek
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - M Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - D Jirák
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| |
Collapse
|
76
|
Lupan AM, Preda MB, Burlacu A. A standard procedure for lentiviral-mediated labeling of murine mesenchymal stromal cells in vitro. Biotechnol Appl Biochem 2019; 66:643-653. [PMID: 31087689 DOI: 10.1002/bab.1765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/13/2019] [Indexed: 12/29/2022]
Abstract
Tracking of stem cells after transplantation is effectively performed in vivo with imaging systems, assuming the cells are adequately labeled to facilitate their recognition. This study aimed to optimize a protocol for fluorescent labeling of mesenchymal stromal cells (MSCs) in vitro, by using a third-generation lentiviral system. Basically, 293T cells are seeded in high-glucose Dulbecco's modified Eagle medium with 10% FBS one day before transfection. Transfection is done for 24 h using a mix of transfer, packaging, regulatory, and envelope plasmids, in molar ratio of 4:2:1:1, respectively. After transfection, the cells are further cultured for two days. During this period, the viral medium is harvested two times, at 24-h intervals, with the first round being stored at 4°C until the second round is completed. The pooled viral medium is frozen in single-use aliquots. MSCs are transduced with 25 multiplicity of infection (MOI) and one day later the cells are passaged at standard seeding density and further grown for three days, when the fluorescence reach the maximum level. Our protocol provides particular experimental details for permanent MSC labeling that makes the procedure highly effective for therapeutic purposes, without affecting the functional properties of stem cells.
Collapse
Affiliation(s)
- Ana-Mihaela Lupan
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu", Bucharest, Romania
| | - Mihai Bogdan Preda
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu", Bucharest, Romania
| | - Alexandrina Burlacu
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
77
|
Yan Y, Shi P, Song W, Bi S. Chemiluminescence and Bioluminescence Imaging for Biosensing and Therapy: In Vitro and In Vivo Perspectives. Theranostics 2019; 9:4047-4065. [PMID: 31281531 PMCID: PMC6592176 DOI: 10.7150/thno.33228] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
Chemiluminescence (CL) and bioluminescence (BL) imaging technologies, which require no external light source so as to avoid the photobleaching, background interference and autoluminescence, have become powerful tools in biochemical analysis and biomedical science with the development of advanced imaging equipment. CL imaging technology has been widely applied to high-throughput detection of a variety of analytes because of its high sensitivity, high efficiency and high signal-to-noise ratio (SNR). Using luciferase and fluorescent proteins as reporters, various BL imaging systems have been developed innovatively for real-time monitoring of diverse molecules in vivo based on the reaction between luciferin and the substrate. Meanwhile, the kinetics of protein interactions even in deep tissues has been studied by BL imaging. In this review, we summarize in vitro and in vivo applications of CL and BL imaging for biosensing and therapy. We first focus on in vitro CL imaging from the view of improving the sensitivity. Then, in vivo CL applications in cells and tissues based on different CL systems are demonstrated. Subsequently, the recent in vitro and in vivo applications of BL imaging are summarized. Finally, we provide the insight into the development trends and future perspectives of CL and BL imaging technologies.
Collapse
|
78
|
Filippi M, Garello F, Pasquino C, Arena F, Giustetto P, Antico F, Terreno E. Indocyanine green labeling for optical and photoacoustic imaging of mesenchymal stem cells after in vivo transplantation. JOURNAL OF BIOPHOTONICS 2019; 12:e201800035. [PMID: 30471202 DOI: 10.1002/jbio.201800035] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 06/09/2023]
Abstract
The transplantation of mesenchymal stem cells (MSCs) holds great promise for the treatment of a plethora of human diseases, but new noninvasive procedures are needed to monitor the cell fate in vivo. Already largely used in medical diagnostics, the fluorescent dye indocyanine green (ICG) is an established dye to track limited numbers of cells by optical imaging (OI), but it can also be visualized by photoacoustic imaging (PAI), which provides a higher spatial resolution than pure near infrared fluorescence imaging (NIRF). Because of its successful use in clinical and preclinical examinations, we chose ICG as PAI cell labeling agent. Optimal incubation conditions were defined for an efficient and clinically translatable MSC labeling protocol, such that no cytotoxicity or alterations of the phenotypic profile were observed, and a consistent intracellular uptake of the molecule was achieved. Suspensions of ICG-labeled cells were both optically and optoacoustically detected in vitro, revealing a certain variability in the photoacoustic spectra acquired by varying the excitation wavelength from 680 to 970 nm. Intramuscular engraftments of ICG-labeled MSCs were clearly visualized by both PAI and NIRF over few days after transplantation in the hindlimb of healthy mice, suggesting that the proposed technique retains a considerable potential in the field of transplantation-focused research and therapy. Stem cells were labeled with the Food and Drug Administration (FDA)-approved fluorescent dye ICG, and detected by both PAI and OI, enabling to monitor the cell fate safely, in dual modality, and with good sensitivity and improved spatial resolution.
Collapse
Affiliation(s)
- Miriam Filippi
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Chiara Pasquino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Francesca Arena
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Pierangela Giustetto
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Federica Antico
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | | |
Collapse
|
79
|
Listeria innocua Dps as a nanoplatform for bioluminescence based photodynamic therapy utilizing Gaussia princeps luciferase and zinc protoporphyrin IX. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 20:102005. [PMID: 31048084 PMCID: PMC6712498 DOI: 10.1016/j.nano.2019.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/31/2022]
Abstract
Listeria innocua DNA binding protein from starved cells (LiDps) belongs to the ferritin family and provides a promising self-assembling spherical 12-mer protein scaffold for the generation of functional nanomaterials. We report the creation of a Gaussia princeps luciferase (Gluc)-LiDps fusion protein, with chemical conjugation of Zinc (II)-protoporphyrin IX (ZnPP) to lysine residues on the fusion protein (giving Gluc-LiDps-ZnPP). The Gluc-LiDps-ZnPP conjugate is shown to generate reactive oxygen species (ROS) via Bioluminescence Resonance Energy Transfer (BRET) between the Gluc (470-490 nm) and ZnPP. In vitro, Gluc-LiDps-ZnPP is efficiently taken up by tumorigenic cells (SKBR3 and MDA-MB-231 breast cancer cells). In the presence of coelenterazine, this construct inhibits the proliferation of SKBR3 due to elevated ROS levels. Following exposure to Gluc-LiDps-ZnPP, migration of surviving SKBR3 cells is significantly suppressed. These results demonstrate the potential of the Gluc-LiDps-ZnPP conjugate as a platform for future development of an anticancer photodynamic therapy agent.
Collapse
|
80
|
Lee H, Lee HW, La Lee Y, Jeon YH, Jeong SY, Lee SW, Lee J, Ahn BC. Optimization of Dendritic Cell-Mediated Cytotoxic T-Cell Activation by Tracking of Dendritic Cell Migration Using Reporter Gene Imaging. Mol Imaging Biol 2019; 20:398-406. [PMID: 29027077 DOI: 10.1007/s11307-017-1127-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The aim of this study is to optimize the dendritic cell (DC)-mediated T-cell activation using reporter gene imaging and flow cytometric analysis in living mice. PROCEDURES A murine dendritic cell line (DC2.4) co-expressing effluc and Thy1.1 genes were established by transfection with retroviral vectors. Thy1.1 positive cells were sorted by magnetic bead separation system (DC2.4/effluc). Cell proliferation assay and phenotype analysis to determine the effects of gene transduction on the function of dendritic cells between parental DC2.4 and DC2.4/effluc were performed. To optimize the DC-mediated immune response by cell number or frequency, different cell numbers (5 × 105, 1 × 106, and 2 × 106 DC2.4/effluc) or different frequencies of DC2.4/effluc (first, second, and third injections) were injected in the right footpad of mice. The migration of the DC2.4/effluc into the draining popliteal lymph node of mice was monitored by bioluminescence imaging (BLI). Flow cytometric analysis was performed with splenocytes to determine the cytotoxic T-cell population after injection of DC2.4/effluc. RESULTS Parental DC2.4 and DC2.4/effluc exhibit no significant differences in their proliferation and phenotype. BLI signals were observed in the draining popliteal lymph node at day 1 after injection of DC2.4/effluc in 1 × 106 and 2 × 106 cells-injected groups. The highest BLI signal intensity was detected in 2 × 106 cells-injected mice. On day 11, the BLI signal was detected in only 2 × 106 cell-injected group but not in other groups. Optimized cell numbers (2 × 106) were injected in three animal groups with a different frequency (first, second, and third injection groups). The BLI signal was detected at day 1 and maintained until day 7 in the first injection group, but there is low signal intensity in the second and the third injection groups. Although the expression levels of Thy1.1 gene in the first injection group were very high, there reveals no expression of Thy1.1 gene in the second and the third injection groups. The number of tumor-specific CD8+ T-cells in the spleen significantly increased, as the number of DC injections increases. CONCLUSIONS Successful optimization of DC-mediated cytotoxic T-cell activation in living mice using reporter gene imaging and flow cytometric analysis was achieved. The optimization of DC-mediated cytotoxic T-cell activation could be applied for the future DC-based immunotherapy.
Collapse
Affiliation(s)
- Hongje Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.,Department of Nuclear Medicine, Dongnam Institution of Radiological & Medical Sciences (DIRAMS), Busan, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - You La Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.,Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.
| |
Collapse
|
81
|
Enhanced noninvasive imaging of oncology models using the NIS reporter gene and bioluminescence imaging. Cancer Gene Ther 2019; 27:179-188. [PMID: 30674994 PMCID: PMC7170803 DOI: 10.1038/s41417-019-0081-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/11/2018] [Accepted: 12/28/2018] [Indexed: 02/08/2023]
Abstract
Noninvasive bioluminescence imaging (BLI) of luciferase-expressing tumor cells has advanced pre-clinical evaluation of cancer therapies. Yet despite its successes, BLI is limited by poor spatial resolution and signal penetration, making it unusable for deep tissue or large animal imaging and preventing precise anatomical localization or signal quantification. To refine pre-clinical BLI methods and circumvent these limitations, we compared and ultimately combined BLI with tomographic, quantitative imaging of the sodium iodide symporter (NIS). To this end, we generated tumor cell lines expressing luciferase, NIS, or both reporters, and established tumor models in mice. BLI provided sensitive early detection of tumors and relatively easy monitoring of disease progression. However, spatial resolution was poor, and as the tumors grew, deep thoracic tumor signals were massked by overwhelming surface signals from superficial tumors. In contrast, NIS-expressing tumors were readily distinguished and precisely localized at all tissue depths by positron emission tomography (PET) or single photon emission computed tomography (SPECT) imaging. Furthermore, radiotracer uptake for each tumor could be quantitated noninvasively. Ultimately, combining BLI and NIS imaging represented a significant enhancement over traditional BLI, providing more information about tumor size and location. This combined imaging approach should facilitate comprehensive evaluation of tumor responses to given therapies.
Collapse
|
82
|
Lina IA, Ishida W, Liauw JA, Lo SFL, Elder BD, Perdomo-Pantoja A, Theodros D, Witham TF, Holmes C. A mouse model for the study of transplanted bone marrow mesenchymal stem cell survival and proliferation in lumbar spinal fusion. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2018; 28:710-718. [PMID: 30511246 DOI: 10.1007/s00586-018-5839-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/21/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Bone marrow aspirate has been successfully used alongside a variety of grafting materials to clinically augment spinal fusion. However, little is known about the fate of these transplanted cells. Herein, we develop a novel murine model for the in vivo monitoring of implanted bone marrow cells (BMCs) following spinal fusion. METHODS A clinical-grade scaffold was implanted into immune-intact mice undergoing spinal fusion with or without freshly isolated BMCs from either transgenic mice which constitutively express the firefly luciferase gene or syngeneic controls. The in vivo survival, distribution and proliferation of these luciferase-expressing cells was monitored via bioluminescence imaging over a period of 8 weeks and confirmed via immunohistochemistry. MicroCT imaging was performed 8 weeks to assess fusion. RESULTS Bioluminescence imaging indicated transplanted cell survival and proliferation over the first 2 weeks, followed by a decrease in cell numbers, with transplanted cell survival still evident at the end of the study. New bone formation and increased fusion mass volume were observed in mice implanted with cell-seeded scaffolds. CONCLUSIONS By enabling the tracking of transplanted bone marrow-derived cells during spinal fusion in vivo, this mouse model will be integral to developing a deeper understanding of the biological processes underlying spinal fusion in future studies. These slides can be retrieved under Electronic Supplementary Material.
Collapse
Affiliation(s)
- Ioan A Lina
- Department of Otolaryngology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wataru Ishida
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Jason A Liauw
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Sheng-Fu L Lo
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Benjamin D Elder
- Department of Neurological Surgery, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Alexander Perdomo-Pantoja
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Debebe Theodros
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Timothy F Witham
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA
| | - Christina Holmes
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 1550 Orleans St, Rm 2M-51, Baltimore, MD, 21287, USA.
| |
Collapse
|
83
|
Scarfe L, Taylor A, Sharkey J, Harwood R, Barrow M, Comenge J, Beeken L, Astley C, Santeramo I, Hutchinson C, Ressel L, Smythe J, Austin E, Levy R, Rosseinsky MJ, Adams DJ, Poptani H, Park BK, Murray P, Wilm B. Non-invasive imaging reveals conditions that impact distribution and persistence of cells after in vivo administration. Stem Cell Res Ther 2018; 9:332. [PMID: 30486897 PMCID: PMC6264053 DOI: 10.1186/s13287-018-1076-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/23/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022] Open
Abstract
Background Cell-based regenerative medicine therapies are now frequently tested in clinical trials. In many conditions, cell therapies are administered systemically, but there is little understanding of their fate, and adverse events are often under-reported. Currently, it is only possible to assess safety and fate of cell therapies in preclinical studies, specifically by monitoring animals longitudinally using multi-modal imaging approaches. Here, using a suite of in vivo imaging modalities to explore the fate of a range of human and murine cells, we investigate how route of administration, cell type and host immune status affect the fate of administered cells. Methods We applied a unique imaging platform combining bioluminescence, optoacoustic and magnetic resonance imaging modalities to assess the safety of different human and murine cell types by following their biodistribution and persistence in mice following administration into the venous or arterial system. Results Longitudinal imaging analyses (i) suggested that the intra-arterial route may be more hazardous than intravenous administration for certain cell types, (ii) revealed that the potential of a mouse mesenchymal stem/stromal cell (MSC) line to form tumours depended on administration route and mouse strain and (iii) indicated that clinically tested human umbilical cord (hUC)-derived MSCs can transiently and unexpectedly proliferate when administered intravenously to mice. Conclusions In order to perform an adequate safety assessment of potential cell-based therapies, a thorough understanding of cell biodistribution and fate post administration is required. The non-invasive imaging platform used here can expose not only the general organ distribution of these therapies, but also a detailed view of their presence within different organs and, importantly, tumourigenic potential. Our observation that the hUC-MSCs but not the human bone marrow (hBM)-derived MSCs persisted for a period in some animals suggests that therapies with these cells should proceed with caution. Electronic supplementary material The online version of this article (10.1186/s13287-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lauren Scarfe
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Jack Sharkey
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Rachel Harwood
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool, Liverpool, UK
| | - Joan Comenge
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lydia Beeken
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Cai Astley
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Ilaria Santeramo
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Lorenzo Ressel
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, Liverpool, UK
| | | | | | - Raphael Levy
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | | | - Dave J Adams
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow, UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Brian K Park
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| |
Collapse
|
84
|
Srinivasan RC, Kannisto K, Strom SC, Gramignoli R. Evaluation of different routes of administration and biodistribution of human amnion epithelial cells in mice. Cytotherapy 2018; 21:113-124. [PMID: 30409699 DOI: 10.1016/j.jcyt.2018.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 09/28/2018] [Accepted: 10/07/2018] [Indexed: 01/10/2023]
Abstract
Placenta is a non-controversial and promising source of cells for the treatment of several liver diseases. We previously reported that transplanted human amnion epithelial cells (hAECs) differentiate into hepatocyte-like cells, resulting in correction of mouse models of metabolic liver disease or acute hepatic failure. As part of preclinical safety studies, we investigated the distribution of hAECs using two routes of administration to efficiently deliver hAECs to the liver. Optical imaging is commonly used because it can provide fast, high-throughput, whole-body imaging, thus DiR-labeled hAECs were injected into immunodeficient mice, via the spleen or the tail vein. The cell distribution was monitored using an in vivo imaging system over the next 24 h. After splenic injection, the DiR signal was detected in liver and spleen at 1, 3 and 24 h post-transplant. The distribution was confirmed by analysis of human DNA content at 24 h post-transplant and human-specific cytokeratin 8/18 staining. Tail vein infusion resulted in cell engraftment mainly in the lungs, with minimal detection in the liver. Delivery of cells to the portal vein, via the spleen, resulted in efficient delivery of hAECs to the liver, with minimal, off-target distribution to lungs or other organs.
Collapse
Affiliation(s)
- Raghuraman C Srinivasan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Kannisto
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephen C Strom
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
85
|
Lee M, Yang H, Kim D, Yang M, Park TH, Hong S. Human-like smelling of a rose scent using an olfactory receptor nanodisc-based bioelectronic nose. Sci Rep 2018; 8:13945. [PMID: 30224633 PMCID: PMC6141559 DOI: 10.1038/s41598-018-32155-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/03/2018] [Indexed: 01/10/2023] Open
Abstract
We report a strategy for the human-like smelling of a rose scent utilizing olfactory receptor nanodisc (ND)-based bioelectronic nose devices. In this strategy, a floating electrode (FE)-based carbon nanotube (CNT) field effect transistor (FET) was functionalized with human olfactory receptor 1A2 (hOR1A2)-embedded NDs (hOR1A2NDs). The hOR1A2NDs responded to rose scent molecules specifically, which were monitored electrically using the underlying CNT-FET. This strategy allowed us to quantitatively assess the contents of geraniol and citronellol, the main components of a rose scent, as low as 1 fM and 10 fM, respectively. In addition, it enabled us to selectively discriminate a specific rose odorant from other odorants. Significantly, we also demonstrated that the responses of hOR1A2NDs to a rose scent could be strongly enhanced by enhancer materials like a human nose. Furthermore, the method provided a means to quantitatively evaluate rose scent components in real samples such as rose oil. Since our method allows one to quantitatively evaluate general rose scent ingredients just like a human nose, it could be a powerful strategy for versatile basic research and various applications such as fragrance development.
Collapse
Affiliation(s)
- Minju Lee
- Department of Physics and Astronomy and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea
| | - Heehong Yang
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Korea
- Protein Engineering Laboratory, Recombinants Unit, MOGAM Institute for Biomedical Research, Yongin, 16924, Korea
| | - Daesan Kim
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, Korea
| | - Myungjae Yang
- Department of Physics and Astronomy and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Korea.
| | - Seunghun Hong
- Department of Physics and Astronomy and Institute of Applied Physics, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
86
|
Gangadaran P, Li XJ, Kalimuthu SK, Min OJ, Hong CM, Rajendran RL, Lee HW, Zhu L, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. New Optical Imaging Reporter-labeled Anaplastic Thyroid Cancer-Derived Extracellular Vesicles as a Platform for In Vivo Tumor Targeting in a Mouse Model. Sci Rep 2018; 8:13509. [PMID: 30201988 PMCID: PMC6131173 DOI: 10.1038/s41598-018-31998-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles (EVs), originating from multivesicular bodies by invagination of the endosomal membrane, are communication channels between distant cells. They are natural carriers of exogeneous cellular materials and have been exploited as drug delivery carriers in various diseases. Here, we found that tumor cell-derived EVs can be used as efficient targets in tumors by monitoring with an optical reporter system. Anaplastic thyroid cancer (CAL62) cell-derived EVs with Renilla luciferase (Rluc) were used to target CAL62 tumors in a mouse model. Optical imaging revealed that cancer cell-derived EVs (EV-CAL62/Rluc) targeted the original tumor (CAL62) in mice within 30 min after systemic injection. Furthermore, fluorescence imaging revealed that EV-CAL62/Rluc were internalized into CAL62 tumors in the mice. Ex vivo Optical imaging further confirmed the in vivo finding. Here, we successfully monitored the tumor targeting ability of tumor cell-derived EVs by optical imaging. Based on these results, tumor cell-derived EVs are highly effective natural carriers for drug delivery for cancer therapies.
Collapse
Affiliation(s)
- Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Xiu Juan Li
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Radiology, Taian City Central Hospital, Taian, People's Republic of China
| | - Senthil Kumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Oh Ji Min
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.
| |
Collapse
|
87
|
Combes F, Mc Cafferty S, Meyer E, Sanders NN. Off-Target and Tumor-Specific Accumulation of Monocytes, Macrophages and Myeloid-Derived Suppressor Cells after Systemic Injection. Neoplasia 2018; 20:848-856. [PMID: 30025228 PMCID: PMC6076377 DOI: 10.1016/j.neo.2018.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/16/2018] [Accepted: 06/19/2018] [Indexed: 01/26/2023] Open
Abstract
Solid tumors frequently coexist with a degree of local chronic inflammation. Recruited myeloid cells can therefore be considered as interesting vehicles for tumor-targeted delivery of therapeutic agents. Using in vivo imaging, the short-term accumulation of systemically injected monocytes, macrophages and myeloid-derived suppressor cells (MDSCs) was compared in mice bearing fat pad mammary carcinomas. Monocytes and macrophages demonstrated almost identical in vivo and ex vivo distribution patterns with maximal tumor-associated accumulation seen 48 hours after injection that remained stable over the 4-day follow-up period. However, a substantial accumulation of both cell types was also seen in the liver, spleen and lungs albeit decreasing over time in all three locations. The MDSCs exhibited a similar distribution pattern as the monocytes and macrophages, but demonstrated a better relative on-target fraction over time. Overall, our findings highlight off-target cell accumulation as a major obstacle in the use of myeloid cells as vehicles for therapeutic tumor-targeted agents and indicate that their short-term on-target accumulation is mainly of nonspecific nature.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Séan Mc Cafferty
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Evelyne Meyer
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
88
|
Kalimuthu S, Zhu L, Oh JM, Gangadaran P, Lee HW, Baek SH, Rajendran RL, Gopal A, Jeong SY, Lee SW, Lee J, Ahn BC. Migration of mesenchymal stem cells to tumor xenograft models and in vitro drug delivery by doxorubicin. Int J Med Sci 2018; 15:1051-1061. [PMID: 30013447 PMCID: PMC6036160 DOI: 10.7150/ijms.25760] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/01/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) show therapeutic effects in various types of diseases. MSCs have been shown to migrate towards inflamed or cancerous tissues, and visualized after sacrificing the animal. MSCs are able to deliver drugs to target cells, and are an ideal candidate for cancer therapy. The purpose of this study was to track the migration of MSCs in tumor-bearing mice; MSCs were also used as drug delivery vehicles. Human breast cancer cells (MDA-MB-231) and anaplastic thyroid cancer cells (CAL62) were transduced with lentiviral particles, to express the Renilla luciferase and mCherry (mCherry-Rluc) reporter genes. Human bone marrow-derived MSCs were transduced with lentiviral particles, to express the firefly luciferase and enhanced green fluorescence protein (Fluc2-eGFP) reporter genes (MSC/Fluc). Luciferase activity of the transduced cells was measured by bioluminescence imaging (BLI). Further in vitro migration assays were performed to confirm cancer cells conditioned medium dependent MSC and doxorubicin (DOX) treated MSC migration. MSCs were loaded with DOX, and their therapeutic effects against the cancer cells were studied in vitro. In vivo MSC/Fluc migration in mice having thyroid or breast cancer xenografts was evaluated after systemic injection. Rluc activity of CAL62/Rluc (R2=0.911), MDA-MB-231/Rluc (R2=0.934) cells and Fluc activity of MSC/Fluc (R2=0.91) cells increased with increasing cell numbers, as seen by BLI. eGFP expression of MSC/Fluc was confirmed by confocal microscopy. Similar migration potential was observed between MSC/Fluc and naïve MSCs in migration assay. DOX treated MSCs migration was not decreased compared than MSCs. Migration of the systemically injected MSC/Fluc cells into tumor xenografts (thyroid and breast cancer) was visualized in animal models (p<0.05) and confirmed by ex vivo (p<0.05) BLI. Additionally, MSCs delivered DOX to CAL62/Rluc and MDA-MB-231/Rluc cells, thereby decreasing their Rluc activities. In this study, we confirmed the migration of MSCs to tumor sites in cancer xenograft models using both in vivo and ex vivo BLI imaging. DOX-pretreated MSCs showed enhanced cytotoxic effects. Therefore, this noninvasive reporter gene (Fluc2)-based BLI may be useful for visualizing in vivo tracking of MSCs, which can be used as a drug delivery vehicle for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
89
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
90
|
Oh EJ, Lee HW, Kalimuthu S, Kim TJ, Kim HM, Baek SH, Zhu L, Oh JM, Son SH, Chung HY, Ahn BC. In vivo migration of mesenchymal stem cells to burn injury sites and their therapeutic effects in a living mouse model. J Control Release 2018; 279:79-88. [PMID: 29655989 DOI: 10.1016/j.jconrel.2018.04.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising therapeutic strategy for tissue regeneration and repair. In this study, we non-invasively monitored the tracking of MSCs toward burn injury sites using MSCs expressing firefly luciferase (Fluc) gene in living mice, and evaluated the effects of the MSCs at the injury site. Murine MSCs co-expressing Fluc and green fluorescent protein (GFP) were established using a retroviral system (referred to as MSC/Fluc). To evaluate the ability of MSC migration toward burn injury sites, cutaneous burn injury was induced in the dorsal skin of mice. MSC/Fluc was intravenously administrated into the mice model and bioluminescence imaging (BLI) was performed to monitor MSC tracking at designated time points. BLI signals of MSC/Fluc appeared in burn injury lesions at 4 days after the cell injection and then gradually decreased. Immunoblotting analysis was conducted to determine the expression of neovascularization-related genes such as TGF-β1 and VEGF in burnt skin. The levels of TGF-β1 and VEGF were higher in the MSC/Fluc-treated group than in the burn injury group. Our observations suggested that MSCs might assist burn wound healing and that MSCs expressing Fluc could be a useful tool for optimizing MSC-based therapeutic strategies for burn wound healing.
Collapse
Affiliation(s)
- Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea; Cell & Matrix Research Institute, Kyungpook National University, Daegu, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Tae Jung Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea
| | - Hyun Mi Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Seung Hyun Son
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea.
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea.
| |
Collapse
|
91
|
Li Y, Liu M, Cui J, Yang K, Zhao L, Gong M, Wang Y, He Y, He T, Bi Y. Hepa1-6-FLuc cell line with the stable expression of firefly luciferase retains its primary properties with promising bioluminescence imaging ability. Oncol Lett 2018; 15:6203-6210. [PMID: 29616102 PMCID: PMC5876459 DOI: 10.3892/ol.2018.8132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/01/2018] [Indexed: 01/10/2023] Open
Abstract
Reliable animal models are required for the in vivo study of the molecular mechanisms and effects of chemotherapeutic drugs in hepatocarcinoma. In vivo tracing techniques based on firefly luciferase (FLuc) may optimize the non-invasive monitoring of experimental animals. The present study established a murine Hepa1-6-FLuc cell line that stably expressed a retrovirus-delivered FLuc protein gene. The cell morphology, proliferation, migration and invasion ability of Hepa1-6-FLuc cells were the same as that of the Hepa1-6 cells, and thus is suitable to replace Hepa1-6 cells in the construction of hepatocarcinoma animal models. No differences in subcutaneous tumor mass and its pathomorphology from implanted Hepa1-6-FLuc cells were observed compared with Hepa1-6 control tumors. Bioluminescence imaging indicated that the Luc signal of the Hepa1-6-FLuc cells was consistently strengthened with increases in tumor mass; however, the Luc signal of Hepa1-6-AdFLuc became weaker and eventually disappeared during tumor development. Therefore, compared with the transient expression by adenovirus, stable expression of the FLuc gene in Hepa1-6 cells may better reflect cell proliferation and survival in vivo, and provide a reliable source for the establishment of hepatocarcinoma models.
Collapse
Affiliation(s)
- Yasha Li
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Mengnan Liu
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jiejie Cui
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Ke Yang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Li Zhao
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Mengjia Gong
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yi Wang
- Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yun He
- Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Tongchuan He
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China.,Key Laboratory of Pediatrics in Chongqing, International Science and Technology Cooperation Base of Child Development and Critical Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| |
Collapse
|
92
|
Kalimuthu S, Zhu L, Oh JM, Lee HW, Gangadaran P, Rajendran RL, Baek SH, Jeon YH, Jeong SY, Lee SW, Lee J, Ahn BC. Regulated Mesenchymal Stem Cells Mediated Colon Cancer Therapy Assessed by Reporter Gene Based Optical Imaging. Int J Mol Sci 2018; 19:1002. [PMID: 29584688 PMCID: PMC5979455 DOI: 10.3390/ijms19041002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/24/2018] [Accepted: 03/25/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is the most common cancer in both men and women and the second most common cause of cancer-related deaths. Suicide gene-based therapy with suicide gene-transduced mesenchymal stem cells (MSCs) is a promising therapeutic strategy. A tetracycline-controlled Tet-On inducible system used to regulate gene expression may be a useful tool for gene-based therapies. The aim of this study was to develop therapeutic MSCs with a suicide gene that is induced by an artificial stimulus, to validate therapeutic gene expression, and to monitor the MSC therapy for colon cancer using optical molecular imaging. For our study, we designed the Tet-On system using a retroviral vector and developed a response plasmid RetroX-TRE (tetracycline response element) expressing a mutant form of herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Bone marrow-derived MSCs were transduced using a RetroX-Tet3G (Clontech, CA, USA) regulatory plasmid and RetroX-TRE-HSV1-sr39TK-eGFP-IRES-Fluc2, for a system with a Tet-On (MSC-Tet-TK/Fluc2 or MSC-Tet-TK) or without a Tet-On (MSC-TK/Fluc2 or MSC-TK) function. Suicide gene engineered MSCs were co-cultured with colon cancer cells (CT26/Rluc) in the presence of the prodrug ganciclovir (GCV) after stimulation with or without doxycycline (DOX). Treatment efficiency was monitored by assessing Rluc (CT26/Rluc) and Fluc (MSC-Tet-TK and MSC-TK) activity using optical imaging. The bystander effect of therapeutic MSCs was confirmed in CT26/Rluc cells after GCV treatment. Rluc activity in CT26/Rluc cells decreased significantly with GCV treatment of DOX(+) cells (p < 0.05 and 0.01) whereas no significant changes were observed in DOX(-) cells. In addition, Fluc activity in also decreased significantly with DOX(+) MSC-Tet-TK cells, but no signal was observed in DOX(-) cells. In addition, an MSC-TK bystander effect was also confirmed. We assessed therapy with this system in a colon cancer xenograft model (CT26/Rluc). We successfully transduced cells and developed a Tet-On system with the suicide gene HSV1-sr39TK. Our results confirmed the therapeutic efficiency of a suicide gene with the Tet-On system for colon cancer. In addition, our results provide an innovative therapeutic approach using the Tet-On system to eradicate tumors by administration of MSC-Tet-TK cells with DOX and GCV.
Collapse
Affiliation(s)
- Senthilkumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Yong Hyun Jeon
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea.
| |
Collapse
|
93
|
Gangadaran P, Hong CM, Ahn BC. An Update on in Vivo Imaging of Extracellular Vesicles as Drug Delivery Vehicles. Front Pharmacol 2018; 9:169. [PMID: 29541030 PMCID: PMC5835830 DOI: 10.3389/fphar.2018.00169] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are currently being considered as promising drug delivery vehicles. EVs are naturally occurring vesicles that exhibit many characteristics favorable to serve as drug delivery vehicles. In addition, EVs have inherent properties for treatment of cancers and other diseases. For research and clinical translation of use of EVs as drug delivery vehicles, in vivo tracking of EVs is essential. The latest molecular imaging techniques enable the tracking of EVs in living animals. However, each molecular imaging technique has its certain advantages and limitations for the in vivo imaging of EVs; therefore, understanding the molecular imaging techniques is essential to select the most appropriate imaging technology to achieve the desired imaging goal. In this review, we summarize the characteristics of EVs as drug delivery vehicles and the molecular imaging techniques used in visualizing and monitoring EVs in in vivo environments. Furthermore, we provide a perceptual vision of EVs as drug delivery vehicles and in vivo monitoring of EVs using molecular imaging technologies.
Collapse
Affiliation(s)
| | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University and Hospital, Daegu, South Korea
| |
Collapse
|
94
|
Gomes AL, Gilchrist S, Kersemans V, Westcott M, Smart S. Refinement of in vivo optical imaging: Development of a real-time respiration monitoring system. Lab Anim 2018; 52:531-535. [PMID: 29451416 DOI: 10.1177/0023677218757273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In vivo optical imaging enables detection and quantification of light-emitting compounds from the whole body in small animals such as the mouse, but it typically requires the use of anaesthetics for subject immobilisation due to long exposure times. Excessively deep anaesthesia can result in unacceptably compromised physiology, whilst excessively light anaesthesia can result in animals waking up. Here we report a respiratory monitoring setup for an in vivo bioluminescence and fluorescence imaging device which simultaneously allows real-time adaptive control of anaesthesia depth in multiple animals to (i) potentially increase the consistency between animals, (ii) ensure animals are maintained within minimally intrusive, adequate anaesthetic plane and (iii) provide a valuable refinement strategy for a common challenge within animal-based research.
Collapse
Affiliation(s)
- Ana L Gomes
- 1 Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, UK
| | - Stuart Gilchrist
- 1 Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, UK
| | - Veerle Kersemans
- 1 Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, UK
| | | | - Sean Smart
- 1 Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, UK
| |
Collapse
|
95
|
Zhang C, Yuan M, Han G, Gao Y, Tang C, Li X, Du L, Li M. Novel caged luciferin derivatives can prolong bioluminescence imaging in vitro and in vivo. RSC Adv 2018; 8:19596-19599. [PMID: 35540985 PMCID: PMC9080648 DOI: 10.1039/c8ra02312c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/20/2018] [Indexed: 12/30/2022] Open
Abstract
Based on N-cyclobutylaminoluciferin (cybLuc), a set of high and efficient caged bioluminescent derivatives (Clucs) as firefly luciferase pro-substrates has been developed herein. After careful examination, these molecules exhibited low cytotoxicity and prolonged bioluminescence imaging up to 6 h in vitro and in vivo. Importantly, these caged luciferin derivatives have the potential to serve as long-term tracking tools to explore some biological process by using bioluminescent imaging. A set of high and efficient caged luciferin derivatives exhibited low cytotoxicity and prolonged bioluminescence in vitro and in vivo.![]()
Collapse
Affiliation(s)
- Chaochao Zhang
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Mingliang Yuan
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Guangxi Han
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Yuqi Gao
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Chunchao Tang
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Xiang Li
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Lupei Du
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| | - Minyong Li
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (MOE)
- School of Pharmacy
- Shandong University
- Jinan
| |
Collapse
|
96
|
Verdes A, Holford M. Beach to Bench to Bedside: Marine Invertebrate Biochemical Adaptations and Their Applications in Biotechnology and Biomedicine. Results Probl Cell Differ 2018; 65:359-376. [PMID: 30083928 DOI: 10.1007/978-3-319-92486-1_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The ocean covers more than 70% of the surface of the planet and harbors very diverse ecosystems ranging from tropical coral reefs to the deepest ocean trenches, with some of the most extreme conditions of pressure, temperature, and light. Organisms living in these environments have been subjected to strong selective pressures through millions of years of evolution, resulting in a plethora of remarkable adaptations that serve a variety of vital functions. Some of these adaptations, including venomous secretions and light-emitting compounds or ink, represent biochemical innovations in which marine invertebrates have developed novel and unique bioactive compounds with enormous potential for basic and applied research. Marine biotechnology, defined as the application of science and technology to marine organisms for the production of knowledge, goods, and services, can harness the enormous possibilities of these unique bioactive compounds acting as a bridge between biological knowledge and applications. This chapter highlights some of the most exceptional biochemical adaptions found specifically in marine invertebrates and describes the biotechnological and biomedical applications derived from them to improve the quality of human life.
Collapse
Affiliation(s)
- Aida Verdes
- Facultad de Ciencias, Departamento de Biología (Zoología), Universidad Autónoma de Madrid, Madrid, Spain.
- Department of Chemistry, Hunter College Belfer Research Center, City University of New York, New York, NY, USA.
- Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY, USA.
| | - Mandë Holford
- Department of Chemistry, Hunter College Belfer Research Center, City University of New York, New York, NY, USA.
- Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY, USA.
- The Graduate Center, Program in Biology, Chemistry and Biochemistry, City University of New York, New York, NY, USA.
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
97
|
Gangadaran P, Li XJ, Lee HW, Oh JM, Kalimuthu S, Rajendran RL, Son SH, Baek SH, Singh TD, Zhu L, Jeong SY, Lee SW, Lee J, Ahn BC. A new bioluminescent reporter system to study the biodistribution of systematically injected tumor-derived bioluminescent extracellular vesicles in mice. Oncotarget 2017; 8:109894-109914. [PMID: 29299117 PMCID: PMC5746352 DOI: 10.18632/oncotarget.22493] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 10/28/2017] [Indexed: 12/21/2022] Open
Abstract
In vivo biodistribution and fate of extracellular vesicles (EVs) are still largely unknown and require reliable in vivo tracking techniques. In this study, in vivo bioluminescence imaging (BLI) using Renilla luciferase (Rluc) was developed and applied to monitoring of EVs derived from thyroid cancer (CAL-62 cells) and breast cancer (MDA-MB-231) in nude mice after intravenous administration and was compared with a dye-based labeling method for EV derived from CAL-62 cells. The EVs were successfully labeled with Rluc and visualized by BLI in mice. In vivo distribution of the EVs, as measured by BLI, was consistent with the results of ex vivo organ analysis. EV-CAL-62/Rluc showed strong signals at lung followed by liver, spleen & kidney (P < 0.05). EV-MDA-MB-231/Rluc showed strong signals at liver followed by lung, spleen & kidney (P < 0.05). EV-CAL-62/Rluc and EV-MDA-MB-231/Rluc stayed in animal till day 9 and 3, respectively; showed a differential distribution. Spontaneous EV-CAL-62/Rluc shown distributed mostly to lung followed by liver, spleen & kidney. The new BLI system used to show spontaneous distribution of EV-CAL-62/Rluc in subcutaneous CAL-62/Rluc bearing mice. Dye (DiR)-labeled EV-CAL-62/Rluc showed a different distribution in vivo & ex vivo compared to EV-CAL-62/Rluc. Fluorescent signals were predominately detected in the liver (P < 0.05) and spleen (P < 0.05) regions. The bioluminescent EVs developed in this study may be used for monitoring of EVs in vivo. This novel reporter-imaging approach to visualization of EVs in real time is expected to pave the way for monitoring of EVs in EV-based treatments.
Collapse
Affiliation(s)
- Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Xiu Juan Li
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Seung Hyun Son
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Thoudam Debraj Singh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
- Department of Medical Oncology, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi 110029, India
| | - Liya Zhu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| |
Collapse
|
98
|
Ibáñez‐Fonseca A, Ramos TL, González de Torre I, Sánchez‐Abarca LI, Muntión S, Arias FJ, Cañizo MC, Alonso M, Sánchez‐Guijo F, Rodríguez‐Cabello JC. Biocompatibility of two model elastin‐like recombinamer‐based hydrogels formed through physical or chemical cross‐linking for various applications in tissue engineering and regenerative medicine. J Tissue Eng Regen Med 2017; 12:e1450-e1460. [DOI: 10.1002/term.2562] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/08/2017] [Accepted: 08/25/2017] [Indexed: 12/17/2022]
Affiliation(s)
| | - Teresa L. Ramos
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de Salamanca Salamanca Spain
- Unidad de Terapia Celular, Servicio de HematologíaHospital Universitario de Salamanca Salamanca Spain
| | | | - Luis Ignacio Sánchez‐Abarca
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de Salamanca Salamanca Spain
- Unidad de Terapia Celular, Servicio de HematologíaHospital Universitario de Salamanca Salamanca Spain
| | - Sandra Muntión
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de Salamanca Salamanca Spain
- Unidad de Terapia Celular, Servicio de HematologíaHospital Universitario de Salamanca Salamanca Spain
| | | | - María Consuelo Cañizo
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de Salamanca Salamanca Spain
- Unidad de Terapia Celular, Servicio de HematologíaHospital Universitario de Salamanca Salamanca Spain
| | - Matilde Alonso
- BIOFORGE LabUniversity of Valladolid–CIBER‐BBN Valladolid Spain
| | - Fermín Sánchez‐Guijo
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de Salamanca Salamanca Spain
- Unidad de Terapia Celular, Servicio de HematologíaHospital Universitario de Salamanca Salamanca Spain
| | | |
Collapse
|
99
|
Gangadaran P, Rajendran RL, Lee HW, Kalimuthu S, Hong CM, Jeong SY, Lee SW, Lee J, Ahn BC. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J Control Release 2017; 264:112-126. [PMID: 28837823 DOI: 10.1016/j.jconrel.2017.08.022] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/11/2017] [Accepted: 08/19/2017] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) are potential therapies for various diseases, but their angiogenic mechanisms of therapeutic efficacy remain unclear. Here, we describe how MSC-EVs, activates VEGF receptors and downstream angiogenesis pathways. Mouse MSC-EVs were isolated from cell culture medium and characterized using transmission electron microscopy, nanoparticle analysis, and western blotting. In vitro migration, proliferation, and tube formation assays using endothelial cells were used to assess the angiogenic potential of MSC-EVs, and revealed higher levels of cellular migration, proliferation, and tube formation after treatment. qRT-PCR and western blotting (WB) revealed higher protein and mRNA expression of the angiogenic genes VEGFR1 and VEGFR2 in mouse SVEC-4 endothelial cells after MSC-EVs treatment. Additionally, other vital pro-angiogenic pathways (SRC, AKT, and ERK) were activated by in vitro MSC-EV treatment. WB and qRT-PCR revealed enriched presence of VEGF protein and miR-210-3p in MSC-EV. The hindlimb ischemia mouse model was established and MSC-EVs with or without Matrigel (EV-MSC+Gel) were injected into the ischemic area and blood reperfusion was monitored using molecular imaging techniques. The in vivo administration of MSC-EVs increased both blood reperfusion and the formation of new blood vessels in the ischemic limb, with the addition of matrigel enhancing this effect further by releasing EVs slowly. MSC-EVs enhance angiogenesis in ischemic limbs, most likely via the overexpression of VEGFR1 and VEGFR2 in endothelial cells. These findings reveal a novel mechanism of activating receptors by MSC-EVs influence the angiogenesis.
Collapse
Affiliation(s)
- Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 700-721, Republic of Korea.
| |
Collapse
|
100
|
Scarfe L, Brillant N, Kumar JD, Ali N, Alrumayh A, Amali M, Barbellion S, Jones V, Niemeijer M, Potdevin S, Roussignol G, Vaganov A, Barbaric I, Barrow M, Burton NC, Connell J, Dazzi F, Edsbagge J, French NS, Holder J, Hutchinson C, Jones DR, Kalber T, Lovatt C, Lythgoe MF, Patel S, Patrick PS, Piner J, Reinhardt J, Ricci E, Sidaway J, Stacey GN, Starkey Lewis PJ, Sullivan G, Taylor A, Wilm B, Poptani H, Murray P, Goldring CEP, Park BK. Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. NPJ Regen Med 2017; 2:28. [PMID: 29302362 PMCID: PMC5677988 DOI: 10.1038/s41536-017-0029-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/30/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Regenerative medicine therapies hold enormous potential for a variety of currently incurable conditions with high unmet clinical need. Most progress in this field to date has been achieved with cell-based regenerative medicine therapies, with over a thousand clinical trials performed up to 2015. However, lack of adequate safety and efficacy data is currently limiting wider uptake of these therapies. To facilitate clinical translation, non-invasive in vivo imaging technologies that enable careful evaluation and characterisation of the administered cells and their effects on host tissues are critically required to evaluate their safety and efficacy in relevant preclinical models. This article reviews the most common imaging technologies available and how they can be applied to regenerative medicine research. We cover details of how each technology works, which cell labels are most appropriate for different applications, and the value of multi-modal imaging approaches to gain a comprehensive understanding of the responses to cell therapy in vivo.
Collapse
Affiliation(s)
- Lauren Scarfe
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Nathalie Brillant
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - J. Dinesh Kumar
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Noura Ali
- College of Health Science, University of Duhok, Duhok, Iraq
| | - Ahmed Alrumayh
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Mohammed Amali
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Stephane Barbellion
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Vendula Jones
- GlaxoSmithKline, David Jack Centre for Research and Development, Ware, UK
| | - Marije Niemeijer
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Sophie Potdevin
- SANOFI Research and Development, Disposition, Safety and Animal Research, Alfortville, France
| | - Gautier Roussignol
- SANOFI Research and Development, Disposition, Safety and Animal Research, Alfortville, France
| | - Anatoly Vaganov
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Ivana Barbaric
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool, Liverpool, UK
| | | | - John Connell
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Francesco Dazzi
- Department of Haemato-Oncology, King’s College London, London, UK
| | | | - Neil S. French
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Julie Holder
- Roslin Cells, University of Cambridge, Cambridge, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - David R. Jones
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Tammy Kalber
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Cerys Lovatt
- GlaxoSmithKline, David Jack Centre for Research and Development, Ware, UK
| | - Mark F. Lythgoe
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Sara Patel
- ReNeuron Ltd, Pencoed Business Park, Pencoed, Bridgend, UK
| | - P. Stephen Patrick
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Jacqueline Piner
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | | | - Emanuelle Ricci
- Institute of Veterinary Science, University of Liverpool, Liverpool, UK
| | | | - Glyn N. Stacey
- UK Stem Cell Bank, Division of Advanced Therapies, National Institute for Biological Standards Control, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Philip J. Starkey Lewis
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Gareth Sullivan
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Blindern, Oslo, Norway
- Institute of Immunology, Oslo University Hospital-Rikshospitalet, Nydalen, Oslo, Norway
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Blindern, Oslo, Norway
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| |
Collapse
|