1
|
Wang J, Jin C, Cen P, Zhou R, Zhong Y, Tian M, Zhang H. Future direction: molecular imaging-based stem cell research. Eur J Nucl Med Mol Imaging 2025; 52:1614-1617. [PMID: 39800805 DOI: 10.1007/s00259-025-07067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Affiliation(s)
- Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
He XY, Zhou YR, Mu T, Liao YF, Jiang L, Qin Y, Cai JH. Magnetic resonance imaging focused on the ferritin heavy chain 1 reporter gene detects neuronal differentiation in stem cells. Neural Regen Res 2023; 18:1563-1569. [PMID: 36571363 PMCID: PMC10075097 DOI: 10.4103/1673-5374.358608] [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: 11/19/2022] Open
Abstract
The neuronal differentiation of mesenchymal stem cells offers a new strategy for the treatment of neurological disorders. Thus, there is a need to identify a noninvasive and sensitive in vivo imaging approach for real-time monitoring of transplanted stem cells. Our previous study confirmed that magnetic resonance imaging, with a focus on the ferritin heavy chain 1 reporter gene, could track the proliferation and differentiation of bone marrow mesenchymal stem cells that had been transduced with lentivirus carrying the ferritin heavy chain 1 reporter gene. However, we could not determine whether or when bone marrow mesenchymal stem cells had undergone neuronal differentiation based on changes in the magnetic resonance imaging signal. To solve this problem, we identified a neuron-specific enolase that can be differentially expressed before and after neuronal differentiation in stem cells. In this study, we successfully constructed a lentivirus carrying the neuron-specific enolase promoter and expressing the ferritin heavy chain 1 reporter gene; we used this lentivirus to transduce bone marrow mesenchymal stem cells. Cellular and animal studies showed that the neuron-specific enolase promoter effectively drove the expression of ferritin heavy chain 1 after neuronal differentiation of bone marrow mesenchymal stem cells; this led to intracellular accumulation of iron and corresponding changes in the magnetic resonance imaging signal. In summary, we established an innovative magnetic resonance imaging approach focused on the induction of reporter gene expression by a neuron-specific promoter. This imaging method can be used to noninvasively and sensitively detect neuronal differentiation in stem cells, which may be useful in stem cell-based therapies.
Collapse
Affiliation(s)
- Xiao-Ya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yi-Rui Zhou
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tong Mu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing; Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi-Fan Liao
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics; Department of Nuclear Medicine, The Second Hospital of the Army Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jin-Hua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
3
|
Jacobs AH, Schelhaas S, Viel T, Waerzeggers Y, Winkeler A, Zinnhardt B, Gelovani J. Imaging of Gene and Cell-Based Therapies: Basis and Clinical Trials. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
4
|
Madsen SD, Giler MK, Bunnell BA, O'Connor KC. Illuminating the Regenerative Properties of Stem Cells In Vivo with Bioluminescence Imaging. Biotechnol J 2020; 16:e2000248. [PMID: 33089922 DOI: 10.1002/biot.202000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/17/2020] [Indexed: 11/10/2022]
Abstract
Preclinical animal studies are essential to the development of safe and effective stem cell therapies. Bioluminescence imaging (BLI) is a powerful tool in animal studies that enables the real-time longitudinal monitoring of stem cells in vivo to elucidate their regenerative properties. This review describes the application of BLI in preclinical stem cell research to address critical challenges in producing successful stem cell therapeutics. These challenges include stem cell survival, proliferation, homing, stress response, and differentiation. The applications presented here utilize bioluminescence to investigate a variety of stem and progenitor cells in several different in vivo models of disease and implantation. An overview of luciferase reporters is provided, along with the advantages and disadvantages of BLI. Additionally, BLI is compared to other preclinical imaging modalities and potential future applications of this technology are discussed in emerging areas of stem cell research.
Collapse
Affiliation(s)
- Sean D Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Margaret K Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Kim C O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
5
|
Haberberger RV, Barry C, Matusica D. Immortalized Dorsal Root Ganglion Neuron Cell Lines. Front Cell Neurosci 2020; 14:184. [PMID: 32636736 PMCID: PMC7319018 DOI: 10.3389/fncel.2020.00184] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Pain is one of the most significant causes of suffering and disability world-wide, and arguably the most burdensome global health challenge. The growing number of patients suffering from chronic pain conditions such as fibromyalgia, complex regional pain syndrome, migraine and irritable bowel syndrome, not only reflect the complexity and heterogeneity of pain types, but also our lack of understanding of the underlying mechanisms. Sensory neurons within the dorsal root ganglia (DRG) have emerged as viable targets for effective chronic pain therapy. However, DRG's contain different classes of primary sensory neurons including pain-associated nociceptive neurons, non-nociceptive temperature sensing, mechanosensory and chemoreceptive neurons, as well as multiple types of immune and endothelial cells. This cell-population heterogeneity makes investigations of individual subgroups of DRG neurons, such as nociceptors, difficult. In attempts to overcome some of these difficulties, a limited number of immortalized DRG-derived cell lines have been generated over the past few decades. In vitro experiments using DRG-derived cell lines have been useful in understanding sensory neuron function. In addition to retaining phenotypic similarities to primary cultured DRG neurons, these cells offer greater suitability for high throughput assays due to ease of culture, maintenance, growth efficiency and cost-effectiveness. For accurate interpretation and translation of results it is critical, however, that phenotypic similarities and differences of DRG-derived cells lines are methodically compared to native neurons. Published reports to date show notable variability in how these DRG-derived cells are maintained and differentiated. Understanding the cellular and molecular differences stemming from different culture methods, is essential to validate past and future experiments, and enable these cells to be used to their full potential. This review describes currently available DRG-derived cell lines, their known sensory and nociceptor specific molecular profiles, and summarize their morphological features related to differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Christine Barry
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Dusan Matusica
- Anatomy & Histology, College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
6
|
Wang J, Zou W, Ma J, Liu J. Biomaterials and Gene Manipulation in Stem Cell-Based Therapies for Spinal Cord Injury. Stem Cells Dev 2019; 28:239-257. [PMID: 30489226 DOI: 10.1089/scd.2018.0169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI), a prominent health issue, represents a substantial portion of the global health care burden. Stem cell-based therapies provide novel solutions for SCI treatment, yet obstacles remain in the form of low survival rate, uncontrolled differentiation, and functional recovery. The application of engineered biomaterials in stem cell therapy provides a physicochemical microenvironment that mimics the stem cell niche, facilitating self-renewal, stem cell differentiation, and tissue reorganization. Nonetheless, external microenvironment support is inadequate, and some obstacles persist, for example, limited sources, gradual aging, and immunogenicity of stem cells. Targeted stem cell gene manipulation could eliminate many of these drawbacks, allowing safer, more effective use under regulation of intrinsic mechanisms. Additionally, through genetic labeling of stem cells, their role in tissue engineering may be elucidated. Therefore, combining stem cell therapy, materials science, and genetic modification technologies may shed light on SCI treatment. Herein, recent advances and advantages of biomaterials and gene manipulation, especially with respect to stem cell-based therapies, are highlighted, and their joint performance in SCI is evaluated. Current technological limitations and perspectives on future directions are then discussed. Although this combination is still in the early stages of development, it is highly likely to substantially contribute to stem cell-based therapies in the foreseeable future.
Collapse
Affiliation(s)
- Jiayi Wang
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Zou
- 3 College of Life Sciences, Liaoning Normal University, Dalian, China.,4 Liaoning Key Laboratories of Biotechnology and Molecular Drug Research & Development, Dalian, China
| | - Jingyun Ma
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Liu
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Dubois VP, Zotova D, Parkins KM, Swick C, Hamilton AM, Kelly JJ, Ronald JA. Safe Harbor Targeted CRISPR-Cas9 Tools for Molecular-Genetic Imaging of Cells in Living Subjects. CRISPR J 2018; 1:440-449. [PMID: 31021241 DOI: 10.1089/crispr.2018.0030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Noninvasive molecular-genetic imaging of cells expressing imaging reporter genes is an invaluable approach for longitudinal monitoring of the biodistribution and viability of cancer cells and cell-based therapies in preclinical models and patients. However, labeling cells with reporter genes often relies on using gene transfer methods that randomly integrate the reporter genes into the genome, which may cause unwanted and serious detrimental effects. To overcome this, we have developed CRISPR-Cas9 tools to edit cells at the adeno-associated virus site 1 (AAVS1) safe harbour with a large donor construct (∼6.3 kilobases) encoding an antibiotic resistance gene and reporter genes for bioluminescence (BLI) and fluorescence imaging. HEK293T cells were transfected with a dual plasmid system encoding the Cas9 endonuclease and an AAVS1-targeted guide RNA in one plasmid, and a donor plasmid encoding a puromycin resistance gene, tdTomato and firefly luciferase flanked by AAVS1 homology arms. Puromycin-resistant clonal cells were isolated and AAVS1 integration was confirmed via PCR and sequencing of the PCR product. In vitro BLI signal correlated well to cell number (R2 = 0.9988; p < 0.05) and was stable over multiple passages. Engineered cells (2.5 × 106) were injected into the left hind flank of nude mice and in vivo BLI was performed on days 0, 7, 14, 21, and 28. BLI signal trended down from day 0 to day 7, but significantly increased by day 28 due to cell growth (p < 0.05). This describes the first CRISPR-Cas9 system for AAVS1 integration of large gene constructs for molecular-genetic imaging of cells in vivo. With further development, including improving editing efficiency, use of clinically relevant reporters, and evaluation in other cell populations that can be readily expanded in culture (e.g., immortalized cells or T cells), this CRISPR-Cas9 reporter gene system could be broadly applied to a number of in vivo cell tracking studies.
Collapse
Affiliation(s)
- Veronica P Dubois
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Darya Zotova
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Katie M Parkins
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada
| | - Connor Swick
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Amanda M Hamilton
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John J Kelly
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John A Ronald
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada.,3 Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
8
|
Willadsen M, Chaise M, Yarovoy I, Zhang AQ, Parashurama N. Engineering molecular imaging strategies for regenerative medicine. Bioeng Transl Med 2018; 3:232-255. [PMID: 30377663 PMCID: PMC6195904 DOI: 10.1002/btm2.10114] [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: 04/10/2018] [Revised: 08/30/2018] [Accepted: 09/01/2018] [Indexed: 12/15/2022] Open
Abstract
The reshaping of the world's aging population has created an urgent need for therapies for chronic diseases. Regenerative medicine offers a ray of hope, and its complex solutions include material, cellular, or tissue systems. We review basics of regenerative medicine/stem cells and describe how the field of molecular imaging, which is based on quantitative, noninvasive, imaging of biological events in living subjects, can be applied to regenerative medicine in order to interrogate tissues in innovative, informative, and personalized ways. We consider aspects of regenerative medicine for which molecular imaging will benefit. Next, genetic and nanoparticle-based cell imaging strategies are discussed in detail, with modalities like magnetic resonance imaging, optical imaging (near infra-red, bioluminescence), raman microscopy, and photoacoustic microscopy), ultrasound, computed tomography, single-photon computed tomography, and positron emission tomography. We conclude with a discussion of "next generation" molecular imaging strategies, including imaging host tissues prior to cell/tissue transplantation.
Collapse
Affiliation(s)
- Matthew Willadsen
- Department of Chemical and Biological Engineering University at Buffalo, State University of New York, Furnas Hall Buffalo New York 14228
| | - Marc Chaise
- Jacobs School of Medicine and Biomedical Sciences University at Buffalo State University of New York 955 Main St., Buffalo, New York 14203
| | - Iven Yarovoy
- Department of Chemical and Biological Engineering University at Buffalo, State University of New York, Furnas Hall Buffalo New York 14228
| | - An Qi Zhang
- Department of Chemical and Biological Engineering University at Buffalo, State University of New York, Furnas Hall Buffalo New York 14228
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering University at Buffalo, State University of New York, Furnas Hall Buffalo New York 14228.,Department of Biomedical Engineering University at Buffalo, State University of New York, Bonner Hall Buffalo New York 14228.,Clinical and Translation Research Center (CTRC) University at Buffalo, State University of New York 875 Ellicott St., Buffalo, New York 14203
| |
Collapse
|
9
|
Volpe A, Man F, Lim L, Khoshnevisan A, Blower J, Blower PJ, Fruhwirth GO. Radionuclide-fluorescence Reporter Gene Imaging to Track Tumor Progression in Rodent Tumor Models. J Vis Exp 2018:57088. [PMID: 29608157 PMCID: PMC5931757 DOI: 10.3791/57088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Metastasis is responsible for most cancer deaths. Despite extensive research, the mechanistic understanding of the complex processes governing metastasis remains incomplete. In vivo models are paramount for metastasis research, but require refinement. Tracking spontaneous metastasis by non-invasive in vivo imaging is now possible, but remains challenging as it requires long-time observation and high sensitivity. We describe a longitudinal combined radionuclide and fluorescence whole-body in vivo imaging approach for tracking tumor progression and spontaneous metastasis. This reporter gene methodology employs the sodium iodide symporter (NIS) fused to a fluorescent protein (FP). Cancer cells are engineered to stably express NIS-FP followed by selection based on fluorescence-activated cell sorting. Corresponding tumor models are established in mice. NIS-FP expressing cancer cells are tracked non-invasively in vivo at the whole-body level by positron emission tomography (PET) using the NIS radiotracer [18F]BF4-. PET is currently the most sensitive in vivo imaging technology available at this scale and enables reliable and absolute quantification. Current methods either rely on large cohorts of animals that are euthanized for metastasis assessment at varying time points, or rely on barely quantifiable 2D imaging. The advantages of the described method are: (i) highly sensitive non-invasive in vivo 3D PET imaging and quantification, (ii) automated PET tracer production, (iii) a significant reduction in required animal numbers due to repeat imaging options, (iv) the acquisition of paired data from subsequent imaging sessions providing better statistical data, and (v) the intrinsic option for ex vivo confirmation of cancer cells in tissues by fluorescence microscopy or cytometry. In this protocol, we describe all steps required for routine NIS-FP-afforded non-invasive in vivo cancer cell tracking using PET/CT and ex vivo confirmation of in vivo results. This protocol has applications beyond cancer research whenever in vivo localization, expansion and long-time monitoring of a cell population is of interest.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Francis Man
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Lindsay Lim
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Alex Khoshnevisan
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Julia Blower
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Philip J Blower
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London;
| |
Collapse
|
10
|
Characterization of neural stem cells modified with hypoxia/neuron-specific VEGF expression system for spinal cord injury. Gene Ther 2017; 25:27-38. [PMID: 29155421 DOI: 10.1038/gt.2017.92] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 09/30/2017] [Accepted: 10/04/2017] [Indexed: 02/01/2023]
Abstract
Spinal cord injury (SCI) is an incurable disease causing an ischemic environment and functional defect, thus a new therapeutic approach is needed for SCI treatment. Vascular endothelial growth factor (VEGF) is a potent therapeutic gene to treat SCI via angiogenesis and neuroprotection, and both tissue-specific gene expression and high gene delivery efficiency are important for successful gene therapy. Here we design the hypoxia/neuron dual-specific gene expression system (pEpo-NSE) and efficient gene delivery platform can be achieved by the combination ex vivo gene therapy with erythropoietin (Epo) enhancer, neuron-specific enolase (NSE) promoter and neural stem cells (NSCs). An in vitro model, NSCs transfected with pEpo-NSE were consistently and selectively overexpressing therapeutic genes in response to neural differentiation and hypoxic conditions. Also, in SCI model, ex vivo gene therapy using pEpo-NSE system with NSCs significantly enhanced gene delivery efficiency compared with pEpo-NSE system gene therapy alone. However, microarray analysis reveals that introducing exogenous pEpo-NSE and VEGF triggers biological pathways in NSCs such as glycolysis and signaling pathways such as Ras and mitogen-activated protein kinase, leading to cell proliferation, differentiation and apoptosis. Collectively, it indicates that the pEpo-NSE gene expression system works stably in NSCs and ex vivo gene therapy using pEpo-NSE system with NSCs improves gene expression efficiency. However, exogenously introduced pEpo-NSE system has an influence on gene expression profiles in NSCs. Therefore, when we consider ex vivo gene therapy for SCI, the effects of changes in gene expression profiles in NSCs on safety should be investigated.
Collapse
|
11
|
Neuron-Specific Fluorescence Reporter-Based Live Cell Tracing for Transdifferentiation of Mesenchymal Stem Cells into Neurons by Chemical Compound. Stem Cells Int 2017; 2017:8452830. [PMID: 28808446 PMCID: PMC5541830 DOI: 10.1155/2017/8452830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Although transdifferentiation of mesenchymal stem cells (MSCs) into neurons increases the possibility of therapeutic use of MSCs for neurodevelopmental disorders, the use of MSCs has the limitation on differentiation efficiency to neuronal lineage and lack of an easy method to monitor the transdifferentiation. In this study, using time-lapse live cell imaging, we assessed the neuronal differentiation of MSCs induced by a small molecule “NHPDQC (N-hydroxy-2-oxo-3-(3-phenylprophyl)-1,2-dihydroquinoxaline-6-carboxamide, C18H17N3O3).” Plasmid vector containing red fluorescence reporter genes under the control of the tubulin α1 (Tα1) promoter (pTα1-DsRed2) traced the neuronal differentiation of MSCs. Two days after NHPDQC treatment, MSCs showed neuron-like phenotype with neurite outgrowth and high expression of neuron-specific markers in more than 95% cells. The fluorescence signals increased in the cytoplasm of pTα1-DsRed2-transfected MSCs after NHPDQC treatment. In vitro monitoring of MSCs along the time courses showed progressive increase of fluorescence till 30 h after treatment, corresponding with the increase in neurite length. We examined an efficient neuronal differentiation of MSCs by NHPDQC alone and monitored the temporal changes of neuronal differentiation by neuron-specific fluorescence reporter along time. This method would help further our understanding of the differentiation of MSCs to produce neurons by simple treatment of small molecule.
Collapse
|
12
|
Tung JK, Berglund K, Gutekunst CA, Hochgeschwender U, Gross RE. Bioluminescence imaging in live cells and animals. NEUROPHOTONICS 2016; 3:025001. [PMID: 27226972 PMCID: PMC4874058 DOI: 10.1117/1.nph.3.2.025001] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/23/2016] [Indexed: 05/13/2023]
Abstract
The use of bioluminescent reporters in neuroscience research continues to grow at a rapid pace as their applications and unique advantages over conventional fluorescent reporters become more appreciated. Here, we describe practical methods and principles for detecting and imaging bioluminescence from live cells and animals. We systematically tested various components of our conventional fluorescence microscope to optimize it for long-term bioluminescence imaging. High-resolution bioluminescence images from live neurons were obtained with our microscope setup, which could be continuously captured for several hours with no signs of phototoxicity. Bioluminescence from the mouse brain was also imaged noninvasively through the intact skull with a conventional luminescence imager. These methods demonstrate how bioluminescence can be routinely detected and measured from live cells and animals in a cost-effective way with common reagents and equipment.
Collapse
Affiliation(s)
- Jack K. Tung
- Georgia Institute of Technology, Coulter Department of Biomedical Engineering, 313 Ferst Drive, Room 2127, Atlanta, Georgia 30332, United States
- Emory University, Department of Neurosurgery, 101 Woodruff Circle, WMRB Rm 6337, Atlanta, Georgia 30322, United States
| | - Ken Berglund
- Emory University, Department of Neurosurgery, 101 Woodruff Circle, WMRB Rm 6337, Atlanta, Georgia 30322, United States
| | - Claire-Anne Gutekunst
- Emory University, Department of Neurosurgery, 101 Woodruff Circle, WMRB Rm 6337, Atlanta, Georgia 30322, United States
| | - Ute Hochgeschwender
- Central Michigan University, College of Medicine and Neuroscience Program, Department of Neuroscience, 1280 S. East Campus Street, Mt. Pleasant, Michigan 48859, United States
| | - Robert E. Gross
- Georgia Institute of Technology, Coulter Department of Biomedical Engineering, 313 Ferst Drive, Room 2127, Atlanta, Georgia 30332, United States
- Emory University, Department of Neurosurgery, 101 Woodruff Circle, WMRB Rm 6337, Atlanta, Georgia 30322, United States
- Address all correspondence to: Robert E. Gross, E-mail:
| |
Collapse
|
13
|
Increased Understanding of Stem Cell Behavior in Neurodegenerative and Neuromuscular Disorders by Use of Noninvasive Cell Imaging. Stem Cells Int 2016; 2016:6235687. [PMID: 26997958 PMCID: PMC4779824 DOI: 10.1155/2016/6235687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Numerous neurodegenerative and neuromuscular disorders are associated with cell-specific depletion in the human body. This imbalance in tissue homeostasis is in healthy individuals repaired by the presence of endogenous stem cells that can replace the lost cell type. However, in most disorders, a genetic origin or limited presence or exhaustion of stem cells impairs correct cell replacement. During the last 30 years, methods to readily isolate and expand stem cells have been developed and this resulted in a major change in the regenerative medicine field as it generates sufficient amount of cells for human transplantation applications. Furthermore, stem cells have been shown to release cytokines with beneficial effects for several diseases. At present however, clinical stem cell transplantations studies are struggling to demonstrate clinical efficacy despite promising preclinical results. Therefore, to allow stem cell therapy to achieve its full potential, more insight in their in vivo behavior has to be achieved. Different methods to noninvasively monitor these cells have been developed and are discussed. In some cases, stem cell monitoring even reached the clinical setting. We anticipate that by further exploring these imaging possibilities and unraveling their in vivo behavior further improvement in stem cell transplantations will be achieved.
Collapse
|
14
|
Micci MA, Boone DR, Parsley MA, Wei J, Patrikeev I, Motamedi M, Hellmich HL. Development of a novel imaging system for cell therapy in the brain. Stem Cell Res Ther 2015; 6:131. [PMID: 26194790 PMCID: PMC4534109 DOI: 10.1186/s13287-015-0129-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/19/2015] [Accepted: 07/09/2015] [Indexed: 01/19/2023] Open
Abstract
Introduction Stem cells have been evaluated as a potential therapeutic approach for several neurological disorders of the central and peripheral nervous system as well as for traumatic brain and spinal cord injury. Currently, the lack of a reliable and safe method to accurately and non-invasively locate the site of implantation and track the migration of stem cells in vivo hampers the development of stem cell therapy and its clinical application. In this report, we present data that demonstrate the feasibility of using the human sodium iodide symporter (hNIS) as a reporter gene for tracking neural stem cells (NSCs) after transplantation in the brain by using single-photon emission tomography/computed tomography (SPECT/CT) imaging. Methods NSCs were isolated from the hippocampus of adult rats (Hipp-NSCs) and transduced with a lentiviral vector containing the hNIS gene. Hipp-NSCs expressing the hNIS (NIS-Hipp-NSCs) were characterized in vitro and in vivo after transplantation in the rat brain and imaged by using technetium-99m (99mTc) and a small rodent SPECT/CT apparatus. Comparisons were made between Hipp-NSCs and NIS-Hipp-NSCs, and statistical analysis was performed by using two-tailed Student’s t test. Results Our results show that the expression of the hNIS allows the repeated visualization of NSCs in vivo in the brain by using SPECT/CT imaging and does not affect the ability of Hipp-NSCs to generate neuronal and glial cells in vitro and in vivo. Conclusions These data support the use of the hNIS as a reporter gene for non-invasive imaging of NSCs in the brain. The repeated, non-invasive tracking of implanted cells will accelerate the development of effective stem cell therapies for traumatic brain injury and other types of central nervous system injury.
Collapse
Affiliation(s)
- Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Debbie R Boone
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Margaret A Parsley
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Jingna Wei
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Igor Patrikeev
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Helen L Hellmich
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
15
|
Abstract
Stem cell based-therapies are novel therapeutic strategies that hold key for developing new treatments for diseases conditions with very few or no cures. Although there has been an increase in the number of clinical trials involving stem cell-based therapies in the last few years, the long-term risks and benefits of these therapies are still unknown. Detailed in vivo studies are needed to monitor the fate of transplanted cells, including their distribution, differentiation, and longevity over time. Advancements in non-invasive cellular imaging techniques to track engrafted cells in real-time present a powerful tool for determining the efficacy of stem cell-based therapies. In this review, we describe the latest approaches to stem cell labeling and tracking using different imaging modalities.
Collapse
Affiliation(s)
- Amit K Srivastava
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, 217 Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205-1832, USA
| | | |
Collapse
|
16
|
Song Y, Xin X, Zhai X, Xia Z, Shen K. Sequential combination therapy with flavopiridol and autocatalytic caspase-3 driven by amplified hTERT promoter synergistically suppresses human ovarian carcinoma growth in vitro and in mice. J Ovarian Res 2014; 7:121. [PMID: 25528169 PMCID: PMC4302516 DOI: 10.1186/s13048-014-0121-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 12/11/2014] [Indexed: 12/15/2022] Open
Abstract
Background Induction of cell apoptosis and regulation of cell cycle are very attractive for treatments of tumors including ovarian carcinoma. Flavopiridol is a potent small molecular cyclin-dependent kinase(cdk) inhibitor, but its antitumor efficacy is not satisfied yet. Caspase-3 play a major role in the transduction of apoptotic signals and the execution of apoptosis in mammalian cells. We have successfully constructed the recombinant adenovirues AdHTVP2G5-rev-casp3 containing autocatalytic caspase-3 (rev-caspase-3) driven by amplified hTERT promoter system (TSTA-hTERTp). In this study, we applied it with flavopiridol to investigate their antitumor effect on ovarian cancer in vitro and in vivo. Methods Cell viabilities were determined using Cell Counting Kit 8 and flow cytometry. RT-PCR and immunoblotting assays were used to detect cellular apoptotic activities. Tumor growth and survival of mice bearing tumors were studied. Results Flavopiridol or AdHTVP2G5-rev-casp3 at low dosage alone was mildly cytotoxic in vitro with a viability rate of 86.5 ± 4.7% for 300 nM flavopiridol and 88.9 ± 5.4% for AdHTVP2G5-rev-casp3 (MOI 20). By contrast, significant synergism of their sequential combination was observed, and the treatment of AdHTVP2G5-rev-casp3 (MOI 20) infection for 72 h, followed by flavopiridol (300 nM) for 48 h, can result in the most synergistic cell death, with cell survival rate and apoptotic rate of 11.6% and 69.7%, respectively. The sequential combination showed synergistic tumor suppression rate of 77.8%, which was significantly higher than that of AdHTVP2G5-rev-casp3 (33.6%) or flavopiridol (40.1%) alone. The mean survival of mice treated with the combination was 286 ± 8 d, which was synergistically longer than that of mice treated with AdHTVP2G5-rev-casp3 (141 ± 14d), flavopiridol (134 ± 10 d) or controls (106 ± 11 d) (P < 0.01). Conclusions The sequential combination of rev-caspase-3 and flavopiridol result in significant synergistic cell killing effects, significant tumor growth suppression and extended survival of mice bearing OVCAR3 cells. The combination should be further explored as a potential clinically useful regimen against ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13048-014-0121-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yue Song
- Department of Obstetrics and Gynecology, ShengJing Hospital, China Medical University, No. 36, Sanhao street, Heping District, Shen yang, 110004, China.
| | - Xing Xin
- Department of Obstetrics and Gynecology, ShengJing Hospital, China Medical University, No. 36, Sanhao street, Heping District, Shen yang, 110004, China.
| | - Xingyue Zhai
- Department of Obstetrics and Gynecology, ShengJing Hospital, China Medical University, No. 36, Sanhao street, Heping District, Shen yang, 110004, China.
| | - Zhijun Xia
- Department of Obstetrics and Gynecology, ShengJing Hospital, China Medical University, No. 36, Sanhao street, Heping District, Shen yang, 110004, China.
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1, Shuai fuyuan Hutong, Dongcheng District, Beijing, China.
| |
Collapse
|
17
|
Aswendt M, Adamczak J, Tennstaedt A. A review of novel optical imaging strategies of the stroke pathology and stem cell therapy in stroke. Front Cell Neurosci 2014; 8:226. [PMID: 25177269 PMCID: PMC4132298 DOI: 10.3389/fncel.2014.00226] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/22/2014] [Indexed: 12/17/2022] Open
Abstract
Transplanted stem cells can induce and enhance functional recovery in experimental stroke. Invasive analysis has been extensively used to provide detailed cellular and molecular characterization of the stroke pathology and engrafted stem cells. But post mortem analysis is not appropriate to reveal the time scale of the dynamic interplay between the cell graft, the ischemic lesion and the endogenous repair mechanisms. This review describes non-invasive imaging techniques which have been developed to provide complementary in vivo information. Recent advances were made in analyzing simultaneously different aspects of the cell graft (e.g., number of cells, viability state, and cell fate), the ischemic lesion (e.g., blood-brain-barrier consistency, hypoxic, and necrotic areas) and the neuronal and vascular network. We focus on optical methods, which permit simple animal preparation, repetitive experimental conditions, relatively medium-cost instrumentation and are performed under mild anesthesia, thus nearly under physiological conditions. A selection of recent examples of optical intrinsic imaging, fluorescence imaging and bioluminescence imaging to characterize the stroke pathology and engrafted stem cells are discussed. Special attention is paid to novel optimal reporter genes/probes for genetic labeling and tracking of stem cells and appropriate transgenic animal models. Requirements, advantages and limitations of these imaging platforms are critically discussed and placed into the context of other non-invasive techniques, e.g., magnetic resonance imaging and positron emission tomography, which can be joined with optical imaging in multimodal approaches.
Collapse
Affiliation(s)
| | | | - Annette Tennstaedt
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, KölnGermany
| |
Collapse
|
18
|
Song Y, Xin X, Xia Z, Zhai X, Shen K. Selective suppression of autocatalytic caspase-3 driven by two-step transcriptional amplified human telomerase reverse transcriptase promoter on ovarian carcinoma growth in vitro and in mice. Oncol Rep 2014; 32:225-34. [PMID: 24858697 DOI: 10.3892/or.2014.3204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/02/2014] [Indexed: 11/06/2022] Open
Abstract
The objective of our study was to construct recombinant adenovirus (rAd) AdHTVP2G5-rev-casp3, which expresses autocatalytic caspase-3 driven by human telomerase reverse transcriptase promoter (hTERTp) with a two-step transcription amplification (TSTA) system and investigate its antitumor effects on ovarian cancer in vitro and in vivo. Fluorescent detection was used to detect EGFP expression in various cells. Cell viabilities were determined using the Cell Counting Kit-8 and flow cytometry. RT-PCR and immunoblotting assays were used to detect cellular apoptotic activities. Tumor growth and survival of tumor-bearing mice were studied. The hTERTp-TSTA system showed the strongest activity in hTERT-positive cancer cells when compared with hTERTp and cytomeglovirus promoter (CMVp). In contrast, it showed no activity in hTERT‑negative HUVECs. AdHTVP2G5‑rev-casp3 markedly suppressed the survival of AO cells in a dose-dependent modality with a viability rate of 17.8 ± 3.5% at an MOI of 70, which was significantly lower than that by AdHT-rev-casp3 and Ad-rev-casp3 (rAds which express rev-caspase-3 driven by hTERTp and CMVp, respectively). In contrast, AdHTVP2G5‑rev-casp3 induced little HUVEC death with a viability rate of 92.7 ± 5.2% at the same MOI. Additionally, AdHTVP2G5-rev-casp3 (MOI=70) caused significant apoptosis in AO cells with an apoptotic rate of 42%. The tumor growth suppression rate of AdHTVP2G5-rev-casp3 was 81.52%, significantly higher than that of AdHT-rev-casp3 (54.94%) or Ad-rev-casp3 (21.35%). AdHTVP2G5-rev-casp3 significantly improved the survival of tumor-bearing mice with little liver damage, with a mean survival of 258 ± 28 days. These results showed that AdHTVP2G5-rev-casp3 caused effective apoptosis with significant tumor selectivity, strongly suppressed tumor growth and improved mouse survival with little liver toxicity. It can be a potent therapeutic agent for tumor targeted treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yue Song
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Xing Xin
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Zhijun Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Xingyue Zhai
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang 110004, P.R. China
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
19
|
Fruhwirth GO, Diocou S, Blower PJ, Ng T, Mullen G. A whole-body dual-modality radionuclide optical strategy for preclinical imaging of metastasis and heterogeneous treatment response in different microenvironments. J Nucl Med 2014; 55:686-94. [PMID: 24604910 PMCID: PMC6205625 DOI: 10.2967/jnumed.113.127480] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Imaging spontaneous cancer cell metastasis or heterogeneous tumor responses to drug treatment in vivo is difficult to achieve. The goal was to develop a new highly sensitive and reliable preclinical longitudinal in vivo imaging model for this purpose, thereby facilitating discovery and validation of anticancer therapies or molecular imaging agents. METHODS The strategy is based on breast cancer cells stably expressing the human sodium iodide symporter (NIS) fused to a red fluorescent protein, thereby permitting radionuclide and fluorescence imaging. Using whole-body nano-SPECT/CT with (99m)TcO4(-), we followed primary tumor growth and spontaneous metastasis in the presence or absence of etoposide treatment. NIS imaging was used to classify organs as small as individual lymph nodes (LNs) to be positive or negative for metastasis, and results were confirmed by confocal fluorescence microscopy. Etoposide treatment efficacy was proven by ex vivo anticaspase 3 staining and fluorescence microscopy. RESULTS In this preclinical model, we found that the NIS imaging strategy outperformed state-of-the-art (18)F-FDG imaging in its ability to detect small tumors (18.5-fold-better tumor-to-blood ratio) and metastases (LN, 3.6-fold) because of improved contrast in organs close to metastatic sites (12- and 8.5-fold-lower standardized uptake value in the heart and kidney, respectively). We applied the model to assess the treatment response to the neoadjuvant etoposide and found a consistent and reliable improvement in spontaneous metastasis detection. Importantly, we also found that tumor cells in different microenvironments responded in a heterogeneous manner to etoposide treatment, which could be determined only by the NIS-based strategy and not by (18)F-FDG imaging. CONCLUSION We developed a new strategy for preclinical longitudinal in vivo cancer cell tracking with greater sensitivity and reliability than (18)F-FDG PET and applied it to track spontaneous and distant metastasis in the presence or absence of genotoxic stress therapy. Importantly, the model provides sufficient sensitivity and dynamic range to permit the reliable assessment of heterogeneous treatment responses in various microenvironments.
Collapse
Affiliation(s)
- Gilbert O. Fruhwirth
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Seckou Diocou
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Philip J. Blower
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Tony Ng
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
| | - Greg Mullen
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| |
Collapse
|
20
|
Minn I, Menezes ME, Sarkar S, Yarlagadda K, Das SK, Emdad L, Sarkar D, Fisher PB, Pomper MG. Molecular-genetic imaging of cancer. Adv Cancer Res 2014; 124:131-69. [PMID: 25287688 PMCID: PMC4339000 DOI: 10.1016/b978-0-12-411638-2.00004-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular-genetic imaging of cancer using nonviral delivery systems has great potential for clinical application as a safe, efficient, noninvasive tool for visualization of various cellular processes including detection of cancer, and its attendant metastases. In recent years, significant effort has been expended in overcoming technical hurdles to enable clinical adoption of molecular-genetic imaging. This chapter will provide an introduction to the components of molecular-genetic imaging and recent advances on each component leading to safe, efficient clinical applications for detecting cancer. Combination with therapy, namely, generating molecular-genetic theranostic constructs, will provide further impetus for clinical translation of this promising technology.
Collapse
Affiliation(s)
- Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Siddik Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA
| | - Keerthi Yarlagadda
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
21
|
In vivo bioluminescence reporter gene imaging for the activation of neuronal differentiation induced by the neuronal activator neurogenin 1 (Ngn1) in neuronal precursor cells. Eur J Nucl Med Mol Imaging 2013; 40:1607-17. [PMID: 23754760 DOI: 10.1007/s00259-013-2457-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 05/03/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Facilitation of the ability of neuronal lineages derived from transplanted stem cells to differentiate is essential to improve the low efficacy of neuronal differentiation in stem cell therapy in vivo. Neurogenin 1 (Ngn1), a basic helix-loop-helix factor, has been used as an activator of neuronal differentiation. In this study, we monitored the in vivo activation of neuronal differentiation by Ngn1 in neuronal precursor cells using neuron-specific promoter-based optical reporters. METHODS The NeuroD promoter coupled with the firefly luciferase reporter system (pNeuroD-Fluc) was used to monitor differentiation in F11 neuronal precursor cells. In vitro luciferase activity was measured and normalized by protein content. The in vivo-jetPEI(TM) system was used for in vivo transgene delivery. The IVIS 100 imaging system was used to monitor in vivo luciferase activity. RESULTS The Ngn1-induced neuronal differentiation of F11 cells generated neurite outgrowth within 2 days of Ngn1 induction. Immunofluorescence staining demonstrated that early and late neuronal marker expression (βIII-tubulin, NeuroD, MAP2, NF-M, and NeuN) was significantly increased at 3 days after treatment with Ngn1. When Ngn1 and the pNeuroD-Fluc vector were cotransfected into F11 cells, we observed an approximately 11-fold increase in the luciferase signal. An in vivo study showed that bioluminescence signals were gradually increased in Ngn1-treated F11 cells for up to 3 days. CONCLUSION In this study, we examined the in vivo tracking of neuronal differentiation induced by Ngn1 using an optical reporter system. This reporter system could be used effectively to monitor the activation efficiency of neuronal differentiation in grafted stem cells treated with Ngn1 for stem cell therapy.
Collapse
|
22
|
Hwang DW, Lee DS. Optical imaging for stem cell differentiation to neuronal lineage. Nucl Med Mol Imaging 2012; 46:1-9. [PMID: 24900026 DOI: 10.1007/s13139-011-0122-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 11/22/2011] [Accepted: 11/24/2011] [Indexed: 01/14/2023] Open
Abstract
In regenerative medicine, the prospect of stem cell therapy holds great promise for the recovery of injured tissues and effective treatment of intractable diseases. Tracking stem cell fate provides critical information to understand and evaluate the success of stem cell therapy. The recent emergence of in vivo noninvasive molecular imaging has enabled assessment of the behavior of grafted stem cells in living subjects. In this review, we provide an overview of current optical imaging strategies based on cell- or tissue-specific reporter gene expression and of in vivo methods to monitor stem cell differentiation into neuronal lineages. These methods use optical reporters either regulated by neuron-specific promoters or containing neuron-specific microRNA binding sites. Both systems revealed dramatic changes in optical reporter imaging signals in cells differentiating into a neuronal lineage. The detection limit of weak promoters or reporter genes can be greatly enhanced by adopting a yeast GAL4 amplification system or an engineering-enhanced luciferase reporter gene. Furthermore, we propose an advanced imaging system to monitor neuronal differentiation during neurogenesis that uses in vivo multiplexed imaging techniques capable of detecting several targets simultaneously.
Collapse
Affiliation(s)
- Do Won Hwang
- Department of Nuclear Medicine, College of Medicine, Seoul National University, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 Korea ; Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, College of Medicine, Seoul National University, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 Korea ; WCU, Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
23
|
Dwyer RM, Ryan J, Havelin RJ, Morris JC, Miller BW, Liu Z, Flavin R, O'Flatharta C, Foley MJ, Barrett HH, Murphy JM, Barry FP, O'Brien T, Kerin MJ. Mesenchymal Stem Cell-mediated delivery of the sodium iodide symporter supports radionuclide imaging and treatment of breast cancer. Stem Cells 2011; 29:1149-57. [PMID: 21608083 DOI: 10.1002/stem.665] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal Stem Cells (MSCs) migrate specifically to tumors in vivo, and coupled with their capacity to bypass immune surveillance, are attractive vehicles for tumor-targeted delivery of therapeutic agents. This study aimed to introduce MSC-mediated expression of the sodium iodide symporter (NIS) for imaging and therapy of breast cancer. Tumor bearing animals received an intravenous or intratumoral injection of NIS expressing MSCs (MSC-NIS), followed by (99m) Technetium pertechnetate imaging 3-14 days later using a BazookaSPECT γ-camera. Tissue was harvested for analysis of human NIS (hNIS) expression by relative quantitative-polymerase chain reaction. Therapy animals received an i.p. injection of (131) I or saline 14 days after injection of MSC-NIS, and tumor volume was monitored for 8 weeks. After injection of MSC-NIS, BazookaSPECT imaging revealed an image of animal intestines and chest area at day 3, along with a visible weak tumor image. By day 14, the tumor was visible with a significant reduction in radionuclide accumulation in nontarget tissue observed. hNIS gene expression was detected in the intestines, heart, lungs, and tumors at early time points but later depleted in nontarget tissues and persisted at the tumor site. Based on imaging/biodistribution data, animals received a therapeutic dose of (131) I 14 days after MSC-NIS injection. This resulted in a significant reduction in tumor growth (mean ± SEM, 236 ± 62 mm(3) vs. 665 ± 204 mm(3) in controls). The ability to track MSC migration and transgene expression noninvasively in real time before therapy is a major advantage to this strategy. This promising data supports the feasibility of this approach as a novel therapy for breast cancer.
Collapse
Affiliation(s)
- Roisin M Dwyer
- Discipline of Surgery, National University of Ireland Galway, Ireland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Cell-based therapies, such as adoptive immunotherapy and stem-cell therapy, have received considerable attention as novel therapeutics in oncological research and clinical practice. The development of effective therapeutic strategies using tumor-targeted cells requires the ability to determine in vivo the location, distribution, and long-term viability of the therapeutic cell populations as well as their biological fate with respect to cell activation and differentiation. In conjunction with various noninvasive imaging modalities, cell-labeling methods, such as exogenous labeling or transfection with a reporter gene, allow visualization of labeled cells in vivo in real time, as well as monitoring and quantifying cell accumulation and function. Such cell-tracking methods also have an important role in basic cancer research, where they serve to elucidate novel biological mechanisms. In this Review, we describe the basic principles of cell-tracking methods, explain various approaches to cell tracking, and highlight recent examples for the application of such methods in animals and humans.
Collapse
|
25
|
Ransohoff KJ, Wu JC. Advances in cardiovascular molecular imaging for tracking stem cell therapy. Thromb Haemost 2010; 104:13-22. [PMID: 20458434 PMCID: PMC3014322 DOI: 10.1160/th09-08-0530] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 12/30/2009] [Indexed: 12/14/2022]
Abstract
The high mortality rate associated with cardiovascular disease is partially due to the lack of proliferative cells in the heart. Without adequate repair following myocardial infarction, progressive dilation can lead to heart failure. Stem cell therapies present one promising option for treating cardiovascular disease, though the specific mechanisms by which they benefit the heart remain unclear. Before stem cell therapies can be used safely in human populations, their biology must be investigated using innovative technologies such as multi-modality molecular imaging. The present review will discuss the basic principles, labelling techniques, clinical applications, and drawbacks associated with four major modalities: radionuclide imaging, magnetic resonance imaging, bioluminescence imaging, and fluorescence imaging.
Collapse
Affiliation(s)
- Katherine J Ransohoff
- Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305-5324, USA.
| | | |
Collapse
|
26
|
Choi Y, Jeon YH, Paik JH, Ko J, Choi DH, Chung JK, Kim CW. In Vivo Scintigraphic Imaging of Antitumor Effects by Combined Radioiodine Therapy and Human Sodium Iodide Symporter Gene Immunotherapy. Mol Imaging 2010. [DOI: 10.2310/7290.2010.00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Yun Choi
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Hyun Jeon
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Ho Paik
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jinkyung Ko
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dae Han Choi
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - June Key Chung
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Chul Woo Kim
- From the Departments of Pathology, Tumor Biology, and Nuclear Medicine; Tumor Immunity Medical Research Centre; and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
27
|
Damia G, D'Incalci M. Contemporary pre-clinical development of anticancer agents--what are the optimal preclinical models? Eur J Cancer 2009; 45:2768-81. [PMID: 19762228 DOI: 10.1016/j.ejca.2009.08.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 08/07/2009] [Indexed: 02/07/2023]
Abstract
The successful identification of novel effective anticancer drugs is largely dependent on the use of appropriate preclinical experimental models that should possibly mimic the complexity of different cancer diseases. The huge number of targets suitable for the design of new anticancer drugs is producing hundreds of novel molecules that require appropriate experimental models to investigate their mode of action and antitumour activity in order to select for clinical investigation the ones with higher chances of being clinically effective. However, our ability to predict the clinical efficacy of a new compound in the clinic based on preclinical data is still limited. This paper overviews the in vitro/in vivo preclinical systems that are currently used to test either compounds with an unknown mechanism of action or compounds designed to hit cancer-specific or cancer-related molecular targets. Examples of experimental models successfully used to identify novel compounds are provided. Xenografts are still the most commonly used in vivo models in drug development due to their high degree of reproducibility and because, in some cases, particularly when orthotopically transplanted, they maintain several biological properties of the human tumours they derive from. Genetic models are very useful for target validation, but are often not sufficiently reproducible to be used for drug evaluation. The variety of animal models can be effectively used to optimally test drugs that presumably act by a defined mode of action, but final success is highly dependent on the ability of drug development teams to integrate different expertises such as biology, chemistry, pharmacology, toxicology and clinical oncology into a clever and well orchestrated plan that keeps in consideration both the complexity of cancer diseases, involving alterations of different pathways, and the complexity of drugs whose pharmacological properties are crucial to obtain the desired effects.
Collapse
Affiliation(s)
- Giovanna Damia
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, Milan 20157, Italy.
| | | |
Collapse
|
28
|
Method of bioluminescence imaging for molecular imaging of physiological and pathological processes. Methods 2009; 48:139-45. [PMID: 19324090 DOI: 10.1016/j.ymeth.2009.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Accepted: 03/11/2009] [Indexed: 01/20/2023] Open
Abstract
Molecular imaging has emerged as a powerful tool in basic, pre-clinical and clinical research for monitoring a variety of molecular and cellular processes in living organisms. Optical imaging techniques, mainly bioluminescence imaging, have extensively been used to study biological processes because of their exquisite sensitivity and high signal-to noise ratio. However, current applications have mainly been limited to small animals due to attenuation and scattering of light by tissues but efforts are ongoing to overcome these hurdles. Here, we focus on bioluminescence imaging by giving a brief overview of recent advances in instrumentation, current available reporter gene-reporter probe systems and applications such as cell trafficking, protein-protein interactions and imaging endogenous processes.
Collapse
|
29
|
Waerzeggers Y, Monfared P, Viel T, Winkeler A, Voges J, Jacobs AH. Methods to monitor gene therapy with molecular imaging. Methods 2009; 48:146-60. [PMID: 19318125 DOI: 10.1016/j.ymeth.2009.03.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 03/11/2009] [Indexed: 01/08/2023] Open
Abstract
Recent progress in scientific and clinical research has made gene therapy a promising option for efficient and targeted treatment of several inherited and acquired disorders. One of the most critical issues for ensuring success of gene-based therapies is the development of technologies for non-invasive monitoring of the distribution and kinetics of vector-mediated gene expression. In recent years many molecular imaging techniques for safe, repeated and high-resolution in vivo imaging of gene expression have been developed and successfully used in animals and humans. In this review molecular imaging techniques for monitoring of gene therapy are described and specific use of these methods in the different steps of a gene therapy protocol from gene delivery to assessment of therapy response is illustrated. Linking molecular imaging (MI) to gene therapy will eventually help to improve the efficacy and safety of current gene therapy protocols for human application and support future individualized patient treatment.
Collapse
Affiliation(s)
- Yannic Waerzeggers
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck Institute for Neurological Research and Faculty of Medicine, University of Cologne, Gleuelerstrasse 50, Cologne 50931, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Arbab AS, Janic B, Haller J, Pawelczyk E, Liu W, Frank JA. In Vivo Cellular Imaging for Translational Medical Research. Curr Med Imaging 2009; 5:19-38. [PMID: 19768136 DOI: 10.2174/157340509787354697] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Personalized treatment using stem, modified or genetically engineered, cells is becoming a reality in the field of medicine, in which allogenic or autologous cells can be used for treatment and possibly for early diagnosis of diseases. Hematopoietic, stromal and organ specific stem cells are under evaluation for cell-based therapies for cardiac, neurological, autoimmune and other disorders. Cytotoxic or genetically altered T-cells are under clinical trial for the treatment of hematopoietic or other malignant diseases. Before using stem cells in clinical trials, translational research in experimental animal models are essential, with a critical emphasis on developing noninvasive methods for tracking the temporal and spatial homing of these cells to target tissues. Moreover, it is necessary to determine the transplanted cell's engraftment efficiency and functional capability. Various in vivo imaging modalities are in use to track the movement and incorporation of administered cells. Tagging cells with reporter genes, fluorescent dyes or different contrast agents transforms them into cellular probes or imaging agents. Recent reports have shown that magnetically labeled cells can be used as cellular magnetic resonance imaging (MRI) probes, demonstrating the cell trafficking to target tissues. In this review, we will discuss the methods to transform cells into probes for in vivo imaging, along with their advantages and disadvantages as well as the future clinical applicability of cellular imaging method and corresponding imaging modality.
Collapse
Affiliation(s)
- Ali S Arbab
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, MI
| | | | | | | | | | | |
Collapse
|
31
|
Kang JH, Chung JK. Molecular-genetic imaging based on reporter gene expression. J Nucl Med 2008; 49 Suppl 2:164S-79S. [PMID: 18523072 DOI: 10.2967/jnumed.107.045955] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Molecular imaging includes proteomic, metabolic, cellular biologic process, and genetic imaging. In a narrow sense, molecular imaging means genetic imaging and can be called molecular-genetic imaging. Imaging reporter genes play a leading role in molecular-genetic imaging. There are 3 major methods of molecular-genetic imaging, based on optical, MRI, and nuclear medicine modalities. For each of these modalities, various reporter genes and probes have been developed, and these have resulted in successful transitions from bench to bedside applications. Each of these imaging modalities has its unique advantages and disadvantages. Fluorescent and bioluminescent optical imaging modalities are simple, less expensive, more convenient, and more user friendly than other imaging modalities. Another advantage, especially of bioluminescence imaging, is its ability to detect low levels of gene expression. MRI has the advantage of high spatial resolution, whereas nuclear medicine methods are highly sensitive and allow data from small-animal imaging studies to be translated to clinical practice. Moreover, multimodality imaging reporter genes will allow us to choose the imaging technologies that are most appropriate for the biologic problem at hand and facilitate the clinical application of reporter gene technologies. Reporter genes can be used to visualize the levels of expression of particular exogenous and endogenous genes and several intracellular biologic phenomena, including specific signal transduction pathways, nuclear receptor activities, and protein-protein interactions. This technique provides a straightforward means of monitoring tumor mass and can visualize the in vivo distributions of target cells, such as immune cells and stem cells. Molecular imaging has gradually evolved into an important tool for drug discovery and development, and transgenic mice with an imaging reporter gene can be useful during drug and stem cell therapy development. Moreover, instrumentation improvements, the identification of novel targets and genes, and imaging probe developments suggest that molecular-genetic imaging is likely to play an increasingly important role in the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Joo Hyun Kang
- Department of Nuclear Medicine, Cancer Research Institute, Tumor Immunity Medical Research Center, College of Medicine, Seoul National University, Seoul, Korea
| | | |
Collapse
|
32
|
Ko MH, Kim S, Hwang DW, Ko HY, Kim YH, Lee DS. Bioimaging of the unbalanced expression of microRNA9 and microRNA9* during the neuronal differentiation of P19 cells. FEBS J 2008; 275:2605-16. [DOI: 10.1111/j.1742-4658.2008.06408.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|