1
|
Tong X, Poramba-Liyanage DW, van Hoolwerff M, Riemers FM, Montilla-Rojo J, Warin J, Salvatori D, Camus A, Meulenbelt I, Ramos YFM, Geijsen N, Tryfonidou MA, Shang P. Isolation and tracing of matrix-producing notochordal and chondrocyte cells using ACAN-2A-mScarlet reporter human iPSC lines. SCIENCE ADVANCES 2024; 10:eadp3170. [PMID: 39441923 PMCID: PMC11498221 DOI: 10.1126/sciadv.adp3170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024]
Abstract
The development of human induced pluripotent stem cell (iPSC)-based regenerative therapies is challenged by the lack of specific cell markers to isolate differentiated cell types and improve differentiation protocols. This issue is particularly critical for notochordal-like cells and chondrocytes, which are crucial in treating back pain and osteoarthritis, respectively. Both cell types produce abundant proteoglycan aggrecan (ACAN), crucial for the extracellular matrix. We generated two human iPSC lines containing an ACAN-2A-mScarlet reporter. The reporter cell lines were validated using CRISPR-mediated transactivation and functionally validated during notochord and cartilage differentiation. The ability to isolate differentiated cell populations producing ACAN enables their enrichment even in the absence of specific cell markers and allows for comprehensive studies and protocol refinement. ACAN's prevalence in various tissues (e.g., cardiac and cerebral) underscores the reporter's versatility as a valuable tool for tracking matrix protein production in diverse cell types, benefiting developmental biology, matrix pathophysiology, and regenerative medicine.
Collapse
Affiliation(s)
- Xiaole Tong
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Deepani W. Poramba-Liyanage
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Marcella van Hoolwerff
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank M. Riemers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Joaquin Montilla-Rojo
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Julie Warin
- Université de Nantes, CHU Nantes, Inserm, CR2TI, 44000 Nantes, France
| | - Daniela Salvatori
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Anne Camus
- Université de Nantes, CHU Nantes, Inserm, CR2TI, 44000 Nantes, France
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Yolande F. M. Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Niels Geijsen
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden node, Leiden, Netherlands
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, Netherlands
| | - Peng Shang
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden node, Leiden, Netherlands
| |
Collapse
|
2
|
Reporter Genes for Brain Imaging Using MRI, SPECT and PET. Int J Mol Sci 2022; 23:ijms23158443. [PMID: 35955578 PMCID: PMC9368793 DOI: 10.3390/ijms23158443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023] Open
Abstract
The use of molecular imaging technologies for brain imaging can not only play an important supporting role in disease diagnosis and treatment but can also be used to deeply study brain functions. Recently, with the support of reporter gene technology, optical imaging has achieved a breakthrough in brain function studies at the molecular level. Reporter gene technology based on traditional clinical imaging modalities is also expanding. By benefiting from the deeper imaging depths and wider imaging ranges now possible, these methods have led to breakthroughs in preclinical and clinical research. This article focuses on the applications of magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), and positron emission tomography (PET) reporter gene technologies for use in brain imaging. The tracking of cell therapies and gene therapies is the most successful and widely used application of these techniques. Meanwhile, breakthroughs have been achieved in the research and development of reporter genes and their imaging probe pairs with respect to brain function research. This paper introduces the imaging principles and classifications of the reporter gene technologies of these imaging modalities, lists the relevant brain imaging applications, reviews their characteristics, and discusses the opportunities and challenges faced by clinical imaging modalities based on reporter gene technology. The conclusion is provided in the last section.
Collapse
|
3
|
Wu AM. Protein Engineering for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00045-4] [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
|
Perlman O, Ito H, Gilad AA, McMahon MT, Chiocca EA, Nakashima H, Farrar CT. Redesigned reporter gene for improved proton exchange-based molecular MRI contrast. Sci Rep 2020; 10:20664. [PMID: 33244130 PMCID: PMC7692519 DOI: 10.1038/s41598-020-77576-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Reporter gene imaging allows for non-invasive monitoring of molecular processes in living cells, providing insights on the mechanisms underlying pathology and therapy. A lysine-rich protein (LRP) chemical exchange saturation transfer (CEST) MRI reporter gene has previously been developed and used to image tumor cells, cardiac viral gene transfer, and oncolytic virotherapy. However, the highly repetitive nature of the LRP reporter gene sequence leads to DNA recombination events and the expression of a range of truncated LRP protein fragments, thereby greatly limiting the CEST sensitivity. Here we report the use of a redesigned LRP reporter (rdLRP), aimed to provide excellent stability and CEST sensitivity. The rdLRP contains no DNA repeats or GC rich regions and 30% less positively charged amino-acids. RT-PCR of cell lysates transfected with rdLRP demonstrated a stable reporter gene with a single distinct band corresponding to full-length DNA. A distinct increase in CEST-MRI contrast was obtained in cell lysates of rdLRP transfected cells and in in vivo LRP expressing mouse brain tumors ([Formula: see text], n = 10).
Collapse
Affiliation(s)
- Or Perlman
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA, 02129, USA
| | - Hirotaka Ito
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Assaf A Gilad
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Michael T McMahon
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - E Antonio Chiocca
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hiroshi Nakashima
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian T Farrar
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA, 02129, USA.
| |
Collapse
|
5
|
Lilley LM, Kamper S, Caldwell M, Chia ZK, Ballweg D, Vistain L, Krimmel J, Mills TA, MacRenaris K, Lee P, Waters EA, Meade TJ. Self-Immolative Activation of β-Galactosidase-Responsive Probes for In Vivo MR Imaging in Mouse Models. Angew Chem Int Ed Engl 2020; 59:388-394. [PMID: 31750611 PMCID: PMC6923588 DOI: 10.1002/anie.201909933] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/29/2019] [Indexed: 12/13/2022]
Abstract
Our lab has developed a new series of self-immolative MR agents for the rapid detection of enzyme activity in mouse models expressing β-galactosidase (β-gal). We investigated two molecular architectures to create agents that detect β-gal activity by modulating the coordination of water to GdIII . The first is an intermolecular approach, wherein we designed several structural isomers to maximize coordination of endogenous carbonate ions. The second involves an intramolecular mechanism for q modulation. We incorporated a pendant coordinating carboxylate ligand with a 2, 4, 6, or 8 carbon linker to saturate ligand coordination to the GdIII ion. This renders the agent ineffective. We show that one agent in particular (6-C pendant carboxylate) is an extremely effective MR reporter for the detection of enzyme activity in a mouse model expressing β-gal.
Collapse
Affiliation(s)
- Laura M Lilley
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Sarah Kamper
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Michael Caldwell
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Zer Keen Chia
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - David Ballweg
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Luke Vistain
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Jeffrey Krimmel
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Teresa Anne Mills
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Keith MacRenaris
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Paul Lee
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Emily Alexandria Waters
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, 60208-3113, USA
| |
Collapse
|
6
|
Lilley LM, Kamper S, Caldwell M, Chia ZK, Ballweg D, Vistain L, Krimmel J, Mills TA, MacRenaris K, Lee P, Waters EA, Meade TJ. Self‐Immolative Activation of β‐Galactosidase‐Responsive Probes for In Vivo MR Imaging in Mouse Models. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201909933] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Laura M. Lilley
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Sarah Kamper
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Michael Caldwell
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Zer Keen Chia
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - David Ballweg
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Luke Vistain
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Jeffrey Krimmel
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Teresa Anne Mills
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Keith MacRenaris
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | - Paul Lee
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
| | | | - Thomas J. Meade
- Departments of Chemistry Molecular Biosciences, Neurobiology, and Radiology Northwestern University Evanston IL 60208-3113 USA
- Center for Advanced Molecular Imaging Northwestern University Evanston IL 60208-3113 USA
| |
Collapse
|
7
|
Krekorian M, Fruhwirth GO, Srinivas M, Figdor CG, Heskamp S, Witney TH, Aarntzen EH. Imaging of T-cells and their responses during anti-cancer immunotherapy. Theranostics 2019; 9:7924-7947. [PMID: 31656546 PMCID: PMC6814447 DOI: 10.7150/thno.37924] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has proven to be an effective approach in a growing number of cancers. Despite durable clinical responses achieved with antibodies targeting immune checkpoint molecules, many patients do not respond. The common denominator for immunotherapies that have successfully been introduced in the clinic is their potential to induce or enhance infiltration of cytotoxic T-cells into the tumour. However, in clinical research the molecules, cells and processes involved in effective responses during immunotherapy remain largely obscure. Therefore, in vivo imaging technologies that interrogate T-cell responses in patients represent a powerful tool to boost further development of immunotherapy. This review comprises a comprehensive analysis of the in vivo imaging technologies that allow the characterisation of T-cell responses induced by anti-cancer immunotherapy, with emphasis on technologies that are clinically available or have high translational potential. Throughout we discuss their respective strengths and weaknesses, providing arguments for selecting the optimal imaging options for future research and patient management.
Collapse
Affiliation(s)
- Massis Krekorian
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Gilbert O. Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Timothy H. Witney
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Erik H.J.G. Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Magdoom KN, Brown A, Rey J, Mareci TH, King MA, Sarntinoranont M. MRI of Whole Rat Brain Perivascular Network Reveals Role for Ventricles in Brain Waste Clearance. Sci Rep 2019; 9:11480. [PMID: 31391474 PMCID: PMC6685961 DOI: 10.1038/s41598-019-44938-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022] Open
Abstract
Investigating the mechanisms by which metabolic wastes are cleared from nervous tissue is important for understanding natural function and the pathophysiology of several neurological disorders including Alzheimer's disease. Recent evidence suggests clearance may be the function of annular spaces around cerebral blood vessels, called perivascular spaces (PVS), through which cerebrospinal fluid (CSF) is transported from the subarachnoid space into brain parenchyma to exchange with interstitial fluid (also known as the glymphatic system). In this work, an MRI-based methodology was developed to reconstruct the PVS network in whole rat brain to better elucidate both PVS uptake and clearance pathways. MR visible tracer (Gd-albumin) was infused in vivo into the CSF-filled lateral ventricle followed by ex vivo high-resolution MR imaging at 17.6 T with an image voxel volume two orders of magnitude smaller than previously reported. Imaged tracer distribution patterns were reconstructed to obtain a more complete brain PVS network. Several PVS connections were repeatedly highlighted across different animals, and new PVS connections between ventricles and different parts of the brain parenchyma were revealed suggesting a possible role for the ventricles as a source or sink for solutes in the brain. In the future, this methodology may be applied to understand changes in the PVS network with disease.
Collapse
Affiliation(s)
- Kulam Najmudeen Magdoom
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA
| | - Alec Brown
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Julian Rey
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA
| | - Thomas H Mareci
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Michael A King
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Veterans Affairs Medical Center, Gainesville, FL, USA
| | - Malisa Sarntinoranont
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
9
|
The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part II). Mol Diagn Ther 2019; 23:27-51. [PMID: 30387041 DOI: 10.1007/s40291-018-0367-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present era of precision medicine sees "cancer" as a consequence of molecular derangements occurring at the commencement of the disease process, with morphological changes happening much later in the process of tumourigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound (US) and magnetic resonance imaging (MRI) play an integral role in the detection of disease at the macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumourigenesis. MFI has the potential to play a key role in heralding the transition from the concept of "one-size-fits-all" treatment to "precision medicine". Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called "radiogenomics", which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning and artificial intelligence, the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, the majority of these require validation before clinical translation is possible. In this two part review, we discuss the systematic collaboration across structural, anatomical and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
|
10
|
Wu MR, Hsiao JK, Liu HM, Huang YY, Tseng YJ, Chou PT, Weng TI, Yang CY. In vivo imaging of insulin-secreting human pancreatic ductal cells using MRI reporter gene technique: A feasibility study. Magn Reson Med 2019; 82:763-774. [PMID: 30957300 DOI: 10.1002/mrm.27749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of in vivo imaging of human pancreatic ductal cells by OATP1B3 reporter gene under MRI. METHODS A human cell line (PANC-1) derived from the pancreatic ductal epithelium was used in this study. After transduction of OATP1B3, the cellular physiological functions and the ability of intracellular uptake of the MRI contrast medium (Gd-EOB-DTPA) were examined. Induced differentiation of the PANC-1 cells into hormone-secreting cells were performed to simulate pancreatic β-like cells. The hormone-secreting cells were implanted into rats and in vivo MRI was evaluated. RESULTS The mRNA and proteins of OATP1B3 were highly expressed. No significant change of cellular physiological functions was found after the expression. After induced differentiation, the hormone secretion capacities of the OATP1B3-expressing PANC-1 cells were confirmed. Intra-cellular uptake of Gd-EOB-DTPA was determined in vitro by inductively coupled plasma mass spectrometry and MRI. In vivo MRI of the OATP1B3-expressing xenograft revealed an increased signal intensity after contrast enhancement. CONCLUSION OATP1B3 can be used as a safe and feasible in vivo MRI gene reporter for human pancreatic ductal cells.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.,Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Jui Tseng
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Te-I Weng
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Yi Yang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Imaging, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
11
|
Boutagy NE, Feher A, Alkhalil I, Umoh N, Sinusas AJ. Molecular Imaging of the Heart. Compr Physiol 2019; 9:477-533. [PMID: 30873600 DOI: 10.1002/cphy.c180007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multimodality cardiovascular imaging is routinely used to assess cardiac function, structure, and physiological parameters to facilitate the diagnosis, characterization, and phenotyping of numerous cardiovascular diseases (CVD), as well as allows for risk stratification and guidance in medical therapy decision-making. Although useful, these imaging strategies are unable to assess the underlying cellular and molecular processes that modulate pathophysiological changes. Over the last decade, there have been great advancements in imaging instrumentation and technology that have been paralleled by breakthroughs in probe development and image analysis. These advancements have been merged with discoveries in cellular/molecular cardiovascular biology to burgeon the field of cardiovascular molecular imaging. Cardiovascular molecular imaging aims to noninvasively detect and characterize underlying disease processes to facilitate early diagnosis, improve prognostication, and guide targeted therapy across the continuum of CVD. The most-widely used approaches for preclinical and clinical molecular imaging include radiotracers that allow for high-sensitivity in vivo detection and quantification of molecular processes with single photon emission computed tomography and positron emission tomography. This review will describe multimodality molecular imaging instrumentation along with established and novel molecular imaging targets and probes. We will highlight how molecular imaging has provided valuable insights in determining the underlying fundamental biology of a wide variety of CVDs, including: myocardial infarction, cardiac arrhythmias, and nonischemic and ischemic heart failure with reduced and preserved ejection fraction. In addition, the potential of molecular imaging to assist in the characterization and risk stratification of systemic diseases, such as amyloidosis and sarcoidosis will be discussed. © 2019 American Physiological Society. Compr Physiol 9:477-533, 2019.
Collapse
Affiliation(s)
- Nabil E Boutagy
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Attila Feher
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Imran Alkhalil
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Nsini Umoh
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Albert J Sinusas
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA.,Yale University School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Brewer KD, Spitler R, Lee KR, Chan AC, Barrozo JC, Wakeel A, Foote CS, Machtaler S, Rioux J, Willmann JK, Chakraborty P, Rice BW, Contag CH, Bell CB, Rutt BK. Characterization of Magneto-Endosymbionts as MRI Cell Labeling and Tracking Agents. Mol Imaging Biol 2018; 20:65-73. [PMID: 28616842 DOI: 10.1007/s11307-017-1093-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Magneto-endosymbionts (MEs) show promise as living magnetic resonance imaging (MRI) contrast agents for in vivo cell tracking. Here we characterize the biomedical imaging properties of ME contrast agents, in vitro and in vivo. PROCEDURES By adapting and engineering magnetotactic bacteria to the intracellular niche, we are creating magneto-endosymbionts (MEs) that offer advantages relative to passive iron-based contrast agents (superparamagnetic iron oxides, SPIOs) for cell tracking. This work presents a biomedical imaging characterization of MEs including: MRI transverse relaxivity (r 2) for MEs and ME-labeled cells (compared to a commercially available iron oxide nanoparticle); microscopic validation of labeling efficiency and subcellular locations; and in vivo imaging of a MDA-MB-231BR (231BR) human breast cancer cells in a mouse brain. RESULTS At 7T, r 2 relaxivity of bare MEs was higher (250 s-1 mM-1) than that of conventional SPIO (178 s-1 mM-1). Optimized in vitro loading of MEs into 231BR cells yielded 1-4 pg iron/cell (compared to 5-10 pg iron/cell for conventional SPIO). r 2 relaxivity dropped by a factor of ~3 upon loading into cells, and was on the same order of magnitude for ME-loaded cells compared to SPIO-loaded cells. In vivo, ME-labeled cells exhibited strong MR contrast, allowing as few as 100 cells to be detected in mice using an optimized 3D SPGR gradient-echo sequence. CONCLUSIONS Our results demonstrate the potential of magneto-endosymbionts as living MR contrast agents. They have r 2 relaxivity values comparable to traditional iron oxide nanoparticle contrast agents, and provide strong MR contrast when loaded into cells and implanted in tissue.
Collapse
Affiliation(s)
- Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | - Ryan Spitler
- Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | | | | | | | | | | | - Steven Machtaler
- Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | - James Rioux
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | - Juergen K Willmann
- Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | | | | | - Christopher H Contag
- Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA
| | | | - Brian K Rutt
- Radiology Department and Molecular Imaging Program (MIPS), Stanford University, Stanford, CA, USA. .,Richard M. Lucas Center for Imaging, Stanford University School of Medicine, The Lucas Expansion, Room PS-064, 1201 Welch Road, Stanford, CA, 94305-5488, USA.
| |
Collapse
|
13
|
Quantifying iron content in magnetic resonance imaging. Neuroimage 2018; 187:77-92. [PMID: 29702183 DOI: 10.1016/j.neuroimage.2018.04.047] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/13/2018] [Accepted: 04/20/2018] [Indexed: 01/19/2023] Open
Abstract
Measuring iron content has practical clinical indications in the study of diseases such as Parkinson's disease, Huntington's disease, ferritinopathies and multiple sclerosis as well as in the quantification of iron content in microbleeds and oxygen saturation in veins. In this work, we review the basic concepts behind imaging iron using T2, T2*, T2', phase and quantitative susceptibility mapping in the human brain, liver and heart, followed by the applications of in vivo iron quantification in neurodegenerative diseases, iron tagged cells and ultra-small superparamagnetic iron oxide (USPIO) nanoparticles.
Collapse
|
14
|
Zinnhardt B, Wiesmann M, Honold L, Barca C, Schäfers M, Kiliaan AJ, Jacobs AH. In vivo imaging biomarkers of neuroinflammation in the development and assessment of stroke therapies - towards clinical translation. Theranostics 2018; 8:2603-2620. [PMID: 29774062 PMCID: PMC5956996 DOI: 10.7150/thno.24128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/31/2018] [Indexed: 01/01/2023] Open
Abstract
Modulation of the inflammatory microenvironment after stroke opens a new avenue for the development of novel neurorestorative therapies in stroke. Understanding the spatio-temporal profile of (neuro-)inflammatory imaging biomarkers in detail thereby represents a crucial factor in the development and application of immunomodulatory therapies. The early integration of quantitative molecular imaging biomarkers in stroke drug development may provide key information about (i) early diagnosis and follow-up, (ii) spatio-temporal drug-target engagement (pharmacodynamic biomarker), (iii) differentiation of responders and non-responders in the patient cohort (inclusion/exclusion criteria; predictive biomarkers), and (iv) the mechanism of action. The use of targeted imaging biomarkers for may thus allow clinicians to decipher the profile of patient-specific inflammatory activity and the development of patient-tailored strategies for immunomodulatory and neuro-restorative therapies in stroke. Here, we highlight the recent developments in preclinical and clinical molecular imaging biomarkers of neuroinflammation (endothelial markers, microglia, MMPs, cell labeling, future developments) in stroke and outline how imaging biomarkers can be used in overcoming current translational roadblocks and attrition in order to advance new immunomodulatory compounds within the clinical pipeline.
Collapse
Affiliation(s)
- Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- EU 7 th FP Programme “Imaging Inflammation in Neurodegenerative Diseases (INMiND)”
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
- Department of Nuclear Medicine, Universitätsklinikum Münster, Münster, Germany
| | - Maximilian Wiesmann
- Department of Anatomy, Radboud university medical center, Donders Institute for Brain, Cognition & Behaviour, Nijmegen, The Netherlands
| | - Lisa Honold
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
| | - Cristina Barca
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- Department of Nuclear Medicine, Universitätsklinikum Münster, Münster, Germany
| | - Amanda J Kiliaan
- Department of Anatomy, Radboud university medical center, Donders Institute for Brain, Cognition & Behaviour, Nijmegen, The Netherlands
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- EU 7 th FP Programme “Imaging Inflammation in Neurodegenerative Diseases (INMiND)”
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
- Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| |
Collapse
|
15
|
Non-invasive detection of adeno-associated viral gene transfer using a genetically encoded CEST-MRI reporter gene in the murine heart. Sci Rep 2018; 8:4638. [PMID: 29545551 PMCID: PMC5854573 DOI: 10.1038/s41598-018-22993-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/05/2018] [Indexed: 01/02/2023] Open
Abstract
Research into gene therapy for heart failure has gained renewed interest as a result of improved safety and availability of adeno-associated viral vectors (AAV). While magnetic resonance imaging (MRI) is standard for functional assessment of gene therapy outcomes, quantitation of gene transfer/expression relies upon tissue biopsy, fluorescence or nuclear imaging. Imaging of gene expression through the use of genetically encoded chemical exchange saturation transfer (CEST)-MRI reporter genes could be combined with clinical cardiac MRI methods to comprehensively probe therapeutic gene expression and subsequent outcomes. The CEST-MRI reporter gene Lysine Rich Protein (LRP) was cloned into an AAV9 vector and either administered systemically via tail vein injection or directly injected into the left ventricular free wall of mice. Longitudinal in vivo CEST-MRI performed at days 15 and 45 after direct injection or at 1, 60 and 90 days after systemic injection revealed robust CEST contrast in myocardium that was later confirmed to express LRP by immunostaining. Ventricular structure and function were not impacted by expression of LRP in either study arm. The ability to quantify and link therapeutic gene expression to functional outcomes can provide rich data for further development of gene therapy for heart failure.
Collapse
|
16
|
Abstract
Immunotherapies include various approaches, ranging from stimulating effector mechanisms to counteracting inhibitory and suppressive mechanisms, and creating a forum for discussing the most effective means of advancing these therapies through imaging is the focus of the newly formed Imaging in Cellular and Immune Therapies (ICIT) interest group within the World Molecular Imaging Society. Efforts are being made in the identification and validation of predictive biomarkers for a number of immunotherapies. Without predictive biomarkers, a considerable number of patients may receive treatments that have no chance of offering a benefit. This will reflect poorly on the field of immunotherapy and will yield false hopes in patients while at the same time contributing to significant cost to the healthcare system. This review summarizes the main strategies in cancer immune and cell-based therapies and discusses recent advances in imaging strategies aimed to improve cancer immunotherapy outcomes.
Collapse
Affiliation(s)
- Vladimir Ponomarev
- Department of Radiology, Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Ave Z-2063, Box 501, New York, NY, 10065, USA.
| |
Collapse
|
17
|
He X, Cai J, Li H, Liu B, Qin Y, Zhong Y, Wang L, Liao Y. In Vivo magnetic resonance imaging of xenografted tumors using FTH1 reporter gene expression controlled by a tet-on switch. Oncotarget 2018; 7:78591-78604. [PMID: 27732930 PMCID: PMC5346662 DOI: 10.18632/oncotarget.12519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022] Open
Abstract
As a promising magnetic resonance imaging (MRI) reporter, ferritin has been used to track cells in vivo; however, its continuous overexpression can be cytotoxic, which restricts its application. In this study, we aimed to develop a switch to turn this genetic reporter “on” or “off” while monitoring cell grafts via MRI. To accomplish this, we genetically modified the ferritin heavy chain (FTH1) with a Tet-On switch and assessed the expression of FTH1 in transduced neuroblastoma cells (SK-N-SH) in vitro and in xenografted tumors in vivo. We found that FTH1 expression induced by doxycycline (Dox) in SK-N-SH-FTH1 cells depended on treatment dose and duration. We successfully detected T2-weighted MRI contrast in cell grafts after switching “on” the reporter gene using Dox, and this contrast disappeared when we switched it “off”. The genetic reporter FTH1 can thus be switched “on” or “off” throughout longitudinal monitoring of cell grafts, limiting expression to when MRI contrast is needed. The controllable imaging system we have developed minimizes risks from constitutive reporter gene overexpression and facilitates tumor cell monitoring in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaoya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Jinhua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Hao Li
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Bo Liu
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Yi Zhong
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Longlun Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Yifan Liao
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| |
Collapse
|
18
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
19
|
Stem Cell Tracing Through MR Molecular Imaging. Tissue Eng Regen Med 2018; 15:249-261. [PMID: 30603551 DOI: 10.1007/s13770-017-0112-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/09/2017] [Accepted: 12/27/2017] [Indexed: 01/12/2023] Open
Abstract
Stem cell therapy opens a new window in medicine to overcome several diseases that remain incurable. It appears such diseases as cardiovascular disorders, brain injury, multiple sclerosis, urinary system diseases, cartilage lesions and diabetes are curable with stem cell transplantation. However, some questions related to stem cell therapy have remained unanswered. Stem cell imaging allows approval of appropriated strategies such as selection of the type and dose of stem cell, and also mode of cell delivery before being tested in clinical trials. MRI as a non-invasive imaging modality provides proper conditions for this aim. So far, different contrast agents such as superparamagnetic or paramagnetic nanoparticles, ultrasmall superparamagnetic nanoparticles, fluorine, gadolinium and some types of reporter genes have been used for imaging of stem cells. The core subject of these studies is to investigate the survival and differentiation of stem cells, contrast agent's toxicity and long term following of transplanted cells. The promising results of in vivo and some clinical trial studies may raise hope for clinical stem cells imaging with MRI.
Collapse
|
20
|
Wu MR, Liu HM, Lu CW, Shen WH, Lin IJ, Liao LW, Huang YY, Shieh MJ, Hsiao JK. Organic anion-transporting polypeptide 1B3 as a dual reporter gene for fluorescence and magnetic resonance imaging. FASEB J 2018; 32:1705-1715. [PMID: 29146731 PMCID: PMC5892727 DOI: 10.1096/fj.201700767r] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Reporter proteins have broad applications in visualizing molecular events at the cellular, tissue and whole-body levels. Transmembrane transporters recognizing specific molecular domains are of particular interest because they enable the migration of signal-source molecules from the extracellular space to the cytoplasm for subsequent application in multimodality imaging. Organic anion-transporting polypeptides (OATPs) have demonstrated their MRI reporter efficacy. We further expanded their use as a dual-modality reporter in MRI and noninvasive in vivo imaging system (IVIS). We overexpressed OATP1B3 in the HT-1080 sarcoma cell line. Both Gd-EOB-DTPA, an MRI contrast agent, and indocyanine green (ICG), a near-infrared fluorescent dye that provides better deep-tissue detection because of its long wavelength, could be delivered to the intracellular space and imaged in a tumor-bearing nude mouse model. Our in vivo dual-imaging reporter system achieved high sensitivity in MRI and observation periods lasting as long as 96 h in IVIS. Because of the superior temporal and spatial resolutions and the clinical availability of both ICG and Gd-EOB-DTPA, this dual-imaging OATP1B3 system will find biomedical use in tumor biology, stem cell trafficking, and tissue engineering.—Wu, M.-R., Liu, H.-M., Lu, C.-W., Shen, W.-H., Lin, I.-J., Liao, L.-W., Huang, Y.-Y., Shieh, M.-J., Hsiao, J.-K. Organic anion-transporting polypeptide 1B3 as a dual reporter gene for fluorescence and magnetic resonance imaging.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan.,Department of Radiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Chen-Wen Lu
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan.,Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Way-Hone Shen
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - I-Jou Lin
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Li-Wen Liao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
21
|
Slavcev RA, Sum CH, St Jean J, Huh H, Nafissi N. Specific Systems for Evaluation. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 110:99-123. [PMID: 30536228 DOI: 10.1007/978-3-319-78259-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Fluorescent-based visualization techniques have long been used to monitor biological activity. This chapter explores the delivery of reporter genes as a means to assay and track activity in biological systems. Bioluminescence is the production of light due to biochemical processes. By encoding genes for bioluminescence, biological processes can be visualized based on gene expression. This chapter also discusses the primary applications of bioluminescence as seen through bioluminescent imaging techniques, flow cytometry, and PCR-based methods of gene detection. These techniques are described in terms of researching gene expression, cancer therapy, and protein interactions.
Collapse
Affiliation(s)
| | - Chi Hong Sum
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Jesse St Jean
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Haein Huh
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Nafiseh Nafissi
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| |
Collapse
|
22
|
Mukherjee A, Davis HC, Ramesh P, Lu GJ, Shapiro MG. Biomolecular MRI reporters: Evolution of new mechanisms. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2017; 102-103:32-42. [PMID: 29157492 PMCID: PMC5726449 DOI: 10.1016/j.pnmrs.2017.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/23/2017] [Accepted: 05/28/2017] [Indexed: 05/08/2023]
Abstract
Magnetic resonance imaging (MRI) is a powerful technique for observing the function of specific cells and molecules inside living organisms. However, compared to optical microscopy, in which fluorescent protein reporters are available to visualize hundreds of cellular functions ranging from gene expression and chemical signaling to biomechanics, to date relatively few such reporters are available for MRI. Efforts to develop MRI-detectable biomolecules have mainly focused on proteins transporting paramagnetic metals for T1 and T2 relaxation enhancement or containing large numbers of exchangeable protons for chemical exchange saturation transfer. While these pioneering developments established several key uses of biomolecular MRI, such as imaging of gene expression and functional biosensing, they also revealed that low molecular sensitivity poses a major challenge for broader adoption in biology and medicine. Recently, new classes of biomolecular reporters have been developed based on alternative contrast mechanisms, including enhancement of spin diffusivity, interactions with hyperpolarized nuclei, and modulation of blood flow. These novel reporters promise to improve sensitivity and enable new forms of multiplexed and functional imaging.
Collapse
Affiliation(s)
- Arnab Mukherjee
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Hunter C Davis
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pradeep Ramesh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - George J Lu
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
23
|
Li X, Hacker M. Molecular imaging in stem cell-based therapies of cardiac diseases. Adv Drug Deliv Rev 2017; 120:71-88. [PMID: 28734900 DOI: 10.1016/j.addr.2017.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/06/2017] [Accepted: 07/16/2017] [Indexed: 12/26/2022]
Abstract
In the past 15years, despite that regenerative medicine has shown great potential for cardiovascular diseases, the outcome and safety of stem cell transplantation has shown controversial results in the published literature. Medical imaging might be useful for monitoring and quantifying transplanted cells within the heart and to serially characterize the effects of stem cell therapy of the myocardium. From the multiple available noninvasive imaging techniques, magnetic resonance imaging and nuclear imaging by positron (PET) or single photon emission computer tomography (SPECT) are the most used clinical approaches to follow the fate of transplanted stem cells in vivo. In this article, we provide a review on the role of different noninvasive imaging modalities and discuss their advantages and disadvantages. We focus on the different in-vivo labeling and reporter gene imaging strategies for stem cell tracking as well as the concept and reliability to use imaging parameters as noninvasive surrogate endpoints for the evaluation of the post-therapeutic outcome.
Collapse
Affiliation(s)
- Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria.
| |
Collapse
|
24
|
Santoso MR, Yang PC. Molecular Imaging of Stem Cells and Exosomes for Myocardial Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9433-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
25
|
Jiang C, Wu D, Haacke EM. Ferritin-EGFP Chimera as an Endogenous Dual-Reporter for Both Fluorescence and Magnetic Resonance Imaging in Human Glioma U251 Cells. ACTA ACUST UNITED AC 2017; 3:1-8. [PMID: 30042970 PMCID: PMC6024424 DOI: 10.18383/j.tom.2015.00181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A unique hybrid protein ferritin–enhanced green fluorescent protein (EGFP) was built to serve as an endogenous dual reporter for both fluorescence and magnetic resonance imaging (MRI). It consists of a human ferritin heavy chain (an iron-storage protein) at the N terminus, a flexible polypeptide in the middle as a linker, and an EGFP at the C terminus. Through antibiotic screening, we established stable human glioma U251 cell strains that expressed ferritin–EGFP under the control of tetracycline. These cells emitted bright green fluorescence and were easily detected by a fluorescent microscope. Ferritin–EGFP overexpression proved effective in triggering obvious intracellular iron accumulation as shown by Prussian blue staining and by MRI. Further, we found that ferritin–EGFP overexpression did not cause proliferation differences between experimental and control group cells when ferritin–EGFP was expressed for <96 hours. Application of this novel ferritin–EGFP chimera has a promising future for combined optical and MRI approaches to study in vivo imaging at a cellular level.
Collapse
Affiliation(s)
- Caihong Jiang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China and
| | - Dongmei Wu
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China and
| | - E Mark Haacke
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China and.,Department of Radiology, Wayne State University, Detroit, Michigan
| |
Collapse
|
26
|
Genetically encoded iron-associated proteins as MRI reporters for molecular and cellular imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [DOI: 10.1002/wnan.1482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 04/18/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023]
|
27
|
Pothayee N, Cummings DM, Schoenfeld TJ, Dodd S, Cameron HA, Belluscio L, Koretsky AP. Magnetic resonance imaging of odorant activity-dependent migration of neural precursor cells and olfactory bulb growth. Neuroimage 2017; 158:232-241. [PMID: 28669915 DOI: 10.1016/j.neuroimage.2017.06.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/03/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Neural progenitors or neuroblasts are produced by precursor cells in the subventricular zone (SVZ) and migrate along the rostral migratory stream (RMS) to the olfactory bulbs (OB) throughout life. In the OB, these adult born neurons either die or replace existing olfactory interneurons, playing a critical role in the stabilization of OB circuitry. Although several aspects of the addition of new neurons into the OB have been studied, it is unclear whether long-distance activity from the OB can regulate the influx of migrating neuroblasts along the RMS. In this study, iron oxide-assisted MRI was used to track the migration of neuroblasts in combination with reversible naris occlusion to manipulate odorant-induced activity. It was found that decreasing olfactory activity led to a decrease in the rate of neuroblast migration along the RMS. Removal of the naris occlusion led to an increase in migratory rate back to control levels, indicating that olfactory activity has regulatory function on neuroblast migration in the RMS. Blocking odorant activity also led to an arrest in OB growth and re-opening the block led to a rapid re-growth returning the bulb size to control levels. Furthermore, pharmacogenetic elimination of the neuroblasts demonstrated that they were required for re-growth of the bulb following sensory deprivation. Together, these results show that sensory activity, neural migration and OB growth are tightly coupled in an interdependent manner.
Collapse
Affiliation(s)
- Nikorn Pothayee
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M Cummings
- Developmental Neural Plasticity Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen Dodd
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Leonardo Belluscio
- Developmental Neural Plasticity Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan P Koretsky
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Mester A, Oltean-Péter B, Rodean I, Opincariu D, Stănescu A, Lázár E, Benedek I, Benedek I, Benedek I. Magnetic Resonance Imaging of Myocardial Function Following Intracoronary and Intramyocardial Stem Cell Injection. JOURNAL OF INTERDISCIPLINARY MEDICINE 2017. [DOI: 10.1515/jim-2017-0053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Stem cell-based therapy is a new therapeutic option that can be used in patients with cardiac diseases caused by myocardial injury. Cardiac magnetic resonance imaging (MRI) is a new noninvasive imaging method with an increasingly widespread indication. The aim of this review was to evaluate the role of cardiac MRI in patients with myocardial infarction undergoing stem cell therapy. We studied the role of MRI in the assessment of myocardial viability, stem cell tracking, assessment of cell survival rate, and monitoring of the long-term effects of stem cell therapy. Based on the current knowledge in this field, this noninvasive, in vivo cardiac imaging technique has a large indication in this group of patients and plays an important role in all stages of stem cell therapy, from the indication to the long-term follow-up of patients.
Collapse
Affiliation(s)
- András Mester
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Balázs Oltean-Péter
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Ioana Rodean
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Diana Opincariu
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Alexandra Stănescu
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Erzsébet Lázár
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Imre Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| |
Collapse
|
29
|
Schilling F, Ros S, Hu DE, D'Santos P, McGuire S, Mair R, Wright AJ, Mannion E, Franklin RJM, Neves AA, Brindle KM. MRI measurements of reporter-mediated increases in transmembrane water exchange enable detection of a gene reporter. Nat Biotechnol 2017; 35:75-80. [PMID: 27918546 PMCID: PMC5230773 DOI: 10.1038/nbt.3714] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 09/29/2016] [Indexed: 12/17/2022]
Abstract
Non-invasive imaging of gene expression can be used to track implanted cells in vivo but often requires the addition of an exogenous contrast agent that may have limited tissue access. We show that the urea transporter (UT-B) can be used as a gene reporter, where reporter expression is detected using 1H MRI measurements of UT-B-mediated increases in plasma membrane water exchange. HEK cells transfected with the reporter showed an increased apparent water exchange rate (AXR), which increased in line with UT-B expression. AXR values measured in vivo, in UT-B-expressing HEK cell xenografts, were significantly higher (about twofold, P < 0.0001), compared with non-expressing controls. Fluorescence imaging of a red fluorescent protein (mStrawberry), co-expressed with UT-B showed that UT-B expression correlated in a linear fashion with AXR. Transduction of rat brain cells in situ with a lentiviral vector expressing UT-B resulted in about a twofold increase in AXR at the site of virus injection.
Collapse
Affiliation(s)
- Franz Schilling
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Susana Ros
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - De-En Hu
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Paula D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Sarah McGuire
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Alan J. Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Elizabeth Mannion
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Robin J. M. Franklin
- Wellcome Trust–Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom
| | - André A. Neves
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Makela AV, Murrell DH, Parkins KM, Kara J, Gaudet JM, Foster PJ. Cellular Imaging With MRI. Top Magn Reson Imaging 2016; 25:177-186. [PMID: 27748707 DOI: 10.1097/rmr.0000000000000101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cellular magnetic resonance imaging (MRI) is an evolving field of imaging with strong translational and research potential. The ability to detect, track, and quantify cells in vivo and over time allows for studying cellular events related to disease processes and may be used as a biomarker for decisions about treatments and for monitoring responses to treatments. In this review, we discuss methods for labeling cells, various applications for cellular MRI, the existing limitations, strategies to address these shortcomings, and clinical cellular MRI.
Collapse
Affiliation(s)
- Ashley V Makela
- *Imaging Research Laboratories, Robarts Research Institute †Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
31
|
Cho IK, Wang S, Mao H, Chan AWS. Genetic engineered molecular imaging probes for applications in cell therapy: emphasis on MRI approach. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:234-261. [PMID: 27766183 PMCID: PMC5069277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Recent advances in stem cell-based regenerative medicine, cell replacement therapy, and genome editing technologies (i.e. CRISPR-Cas 9) have sparked great interest in in vivo cell monitoring. Molecular imaging promises a unique approach to noninvasively monitor cellular and molecular phenomena, including cell survival, migration, proliferation, and even differentiation at the whole organismal level. Several imaging modalities and strategies have been explored for monitoring cell grafts in vivo. We begin this review with an introduction describing the progress in stem cell technology, with a perspective toward cell replacement therapy. The importance of molecular imaging in reporting and assessing the status of cell grafts and their relation to the local microenvironment is highlighted since the current knowledge gap is one of the major obstacles in clinical translation of stem cell therapy. Based on currently available imaging techniques, we provide a brief discussion on the pros and cons of each imaging modality used for monitoring cell grafts with particular emphasis on magnetic resonance imaging (MRI) and the reporter gene approach. Finally, we conclude with a comprehensive discussion of future directions of applying molecular imaging in regenerative medicine to emphasize further the importance of correlating cell graft conditions and clinical outcomes to advance regenerative medicine.
Collapse
Affiliation(s)
- In K Cho
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| | - Silun Wang
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Anthony WS Chan
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| |
Collapse
|
32
|
Artificial MicroRNAs as Novel Secreted Reporters for Cell Monitoring in Living Subjects. PLoS One 2016; 11:e0159369. [PMID: 27442530 PMCID: PMC4956193 DOI: 10.1371/journal.pone.0159369] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 07/02/2016] [Indexed: 01/08/2023] Open
Abstract
Reporter genes are powerful technologies that can be used to directly inform on the fate of transplanted cells in living subjects. Imaging reporter genes are often employed to quantify cell number, location(s), and viability with various imaging modalities. To complement this, reporters that are secreted from cells can provide a low-cost, in vitro diagnostic test to monitor overall cell viability at relatively high frequency without knowing the locations of all cells. Whereas protein-based secretable reporters have been developed, an RNA-based reporter detectable with amplification inherent PCR-based assays has not been previously described. MicroRNAs (miRNAs) are short non-coding RNAs (18–22 nt) that regulate mRNA translation and are being explored as relatively stable blood-based disease biomarkers. We developed an artificial miRNA-based secreted reporter, called Sec-miR, utilizing a coding sequence that is not expressed endogenously and does not have any known vertebrate target. Sec-miR was detectable in both the cells and culture media of transiently transfected cells. Cells stably expressing Sec-miR also reliably secreted it into the culture media. Mice implanted with parental HeLa cells or HeLa cells expressing both Sec-miR and the bioluminescence imaging (BLI) reporter gene Firefly luciferase (FLuc) were monitored over time for tumor volume, FLuc signal via BLI, and blood levels of Sec-miR. Significantly (p<0.05) higher Sec-miR was found in the blood of mice bearing Sec-miR-expressing tumors compared to parental cell tumors at 21 and 28 days after implantation. Importantly, blood Sec-miR reporter levels after day 21 showed a trend towards correlation with tumor volume (R2 = 0.6090; p = 0.0671) and significantly correlated with FLuc signal (R2 = 0.7067; p<0.05). Finally, we could significantly (p<0.01) amplify Sec-miR secretion into the cell media by chaining together multiple Sec-miR copies (4 instead of 1 or 2) within an expression cassette. Overall, we show that a novel complement of BLI together with a unique Sec-miR reporter adds an in vitro RNA-based diagnostic to enhance the monitoring of transplanted cells. While Sec-miR was not as sensitive as BLI for monitoring cell number, it may be more sensitive than clinically-relevant positron emission tomography (PET) reporter assays. Future work will focus on improving cell detectability via improved secretion of Sec-miR reporters from cells and more sensitive detection platforms, as well as, exploring other miRNA sequences to allow multiplexed monitoring of more than one cell population at a time. Continued development may lead to more refined and precise monitoring of cell-based therapies.
Collapse
|
33
|
Gervois P, Wolfs E, Ratajczak J, Dillen Y, Vangansewinkel T, Hilkens P, Bronckaers A, Lambrichts I, Struys T. Stem Cell-Based Therapies for Ischemic Stroke: Preclinical Results and the Potential of Imaging-Assisted Evaluation of Donor Cell Fate and Mechanisms of Brain Regeneration. Med Res Rev 2016; 36:1080-1126. [DOI: 10.1002/med.21400] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Pascal Gervois
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Esther Wolfs
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Jessica Ratajczak
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Yörg Dillen
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tim Vangansewinkel
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Petra Hilkens
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Annelies Bronckaers
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Ivo Lambrichts
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tom Struys
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| |
Collapse
|
34
|
Li C, Hu CJ, Tang B, Yong X, Luo G, Wu YY, Wang SM, Yu ST, Yang SM. MR molecular imaging of tumors based on an optimal hTERT promoter tyrosinase expression system. Oncotarget 2016; 7:42474-42484. [PMID: 27283901 PMCID: PMC5173149 DOI: 10.18632/oncotarget.9888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/13/2016] [Indexed: 12/01/2022] Open
Abstract
The early diagnosis and treatment of tumors is of vital significance to increase patient survival. Therefore, we constructed a lentiviral vector expressing tyrosinase (TYR) driven by an optimized human telomerase reverse transcriptase (hTERT) promoter or a cytomegalovirus(CMV) promoter in the hopes of performing noninvasive and real-time tumor-specific imaging. First, hTERT-TYR and CMV-TYR were constructed to infect cancer cell lines (telomerase-negative cell line: U2OS; telomerase-positive cell lines: SGC-7901, SW480 and HepG2). Subsequently, stable tyrosinase-expressing cell lines were sorted by flow cytometry out of these infected cancer cell lines. Then, the mRNA and protein levels of tyrosinase were analyzed. Thetyrosinase activity, melanin production and ferric ion adsorption were measured followed by an MR scan. Consequently the results showed that tyrosinase was only expressed in telomerase-positive tumor cells infected by hTERT-TYR, whereas tyrosinase was expressed in both telomerase-negative and telomerase-positive tumor cells infected by CMV-TYR. Finally, we performed in vivo tumor MR using a clinical 3T MR scanner and found increased signals at T1W1 from telomerase-positive cells infected by hTERT-TYR, which revealed that MR scanning could distinguish cells with hTERT -positive cells from hTERT-negative cells infected with the optimized lentivirus. The mechanism underlying this effect is that tyrosinase promotes melanin production and ferric ion adsorption only in hTERT-expressing cells. Taken together, these data show that this optimized hTERT promoter-driving tyrosinase expression system might be a useful diagnostic tool for the detection of tumors using MR imaging.
Collapse
Affiliation(s)
- Chuan Li
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China.,Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Gang Luo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yu-Yun Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Su-Min Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Song-Tao Yu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China.,Department of Oncology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
35
|
MRI Reporter Genes for Noninvasive Molecular Imaging. Molecules 2016; 21:molecules21050580. [PMID: 27213309 PMCID: PMC6273230 DOI: 10.3390/molecules21050580] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 01/17/2023] Open
Abstract
Magnetic resonance imaging (MRI) is one of the most important imaging technologies used in clinical diagnosis. Reporter genes for MRI can be applied to accurately track the delivery of cell in cell therapy, evaluate the therapy effect of gene delivery, and monitor tissue/cell-specific microenvironments. Commonly used reporter genes for MRI usually include genes encoding the enzyme (e.g., tyrosinase and β-galactosidase), the receptor on the cells (e.g., transferrin receptor), and endogenous reporter genes (e.g., ferritin reporter gene). However, low sensitivity limits the application of MRI and reporter gene-based multimodal imaging strategies are common including optical imaging and radionuclide imaging. These can significantly improve diagnostic efficiency and accelerate the development of new therapies.
Collapse
|
36
|
Shaikh F, Jacob A, Van Gestel F, Yaghoubi S. Molecular Imaging in Genetic Medicine. Cureus 2016; 8:e565. [PMID: 27186447 PMCID: PMC4866833 DOI: 10.7759/cureus.565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/11/2016] [Indexed: 12/29/2022] Open
Abstract
The field of biomedical imaging has made significant advances in recent times. This includes extremely high-resolution anatomic imaging and functional imaging of physiologic and pathologic processes as well as novel modalities in optical imaging to evaluate molecular features within the cellular environment. The latter has made it possible to image phenotypic markers of various genotypes that are implicated in human development, behavior, and disease. This article discusses the role of molecular imaging in genetic and precision medicine.
Collapse
Affiliation(s)
- Faiq Shaikh
- Imaging Informatics, University of Pittsburgh Medical Center, Pittsburgh, PA. ; Molecular Imaging, Cellsight Technologies, Inc., San Francisco, CA
| | - Ayden Jacob
- Director of Translational Medicine, Nanoaxis LLC, Neuroscientist, Neuro-Nanotech Division, University of California, Department of Bioengineering ; UCSF Department of Interventional Radiology and Oncology
| | | | | |
Collapse
|
37
|
Stem Cell Imaging: Tools to Improve Cell Delivery and Viability. Stem Cells Int 2016; 2016:9240652. [PMID: 26880997 PMCID: PMC4736428 DOI: 10.1155/2016/9240652] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/06/2015] [Accepted: 10/15/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapy (SCT) has shown very promising preclinical results in a variety of regenerative medicine applications. Nevertheless, the complete utility of this technology remains unrealized. Imaging is a potent tool used in multiple stages of SCT and this review describes the role that imaging plays in cell harvest, cell purification, and cell implantation, as well as a discussion of how imaging can be used to assess outcome in SCT. We close with some perspective on potential growth in the field.
Collapse
|
38
|
Zheng B, von See MP, Yu E, Gunel B, Lu K, Vazin T, Schaffer DV, Goodwill PW, Conolly SM. Quantitative Magnetic Particle Imaging Monitors the Transplantation, Biodistribution, and Clearance of Stem Cells In Vivo. Theranostics 2016; 6:291-301. [PMID: 26909106 PMCID: PMC4737718 DOI: 10.7150/thno.13728] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/01/2015] [Indexed: 12/13/2022] Open
Abstract
Stem cell therapies have enormous potential for treating many debilitating diseases, including heart failure, stroke and traumatic brain injury. For maximal efficacy, these therapies require targeted cell delivery to specific tissues followed by successful cell engraftment. However, targeted delivery remains an open challenge. As one example, it is common for intravenous deliveries of mesenchymal stem cells (MSCs) to become entrapped in lung microvasculature instead of the target tissue. Hence, a robust, quantitative imaging method would be essential for developing efficacious cell therapies. Here we show that Magnetic Particle Imaging (MPI), a novel technique that directly images iron-oxide nanoparticle-tagged cells, can longitudinally monitor and quantify MSC administration in vivo. MPI offers near-ideal image contrast, depth penetration, and robustness; these properties make MPI both ultra-sensitive and linearly quantitative. Here, we imaged, for the first time, the dynamic trafficking of intravenous MSC administrations using MPI. Our results indicate that labeled MSC injections are immediately entrapped in lung tissue and then clear to the liver within one day, whereas standard iron oxide particle (Resovist) injections are immediately taken up by liver and spleen. Longitudinal MPI-CT imaging also indicated a clearance half-life of MSC iron oxide labels in the liver at 4.6 days. Finally, our ex vivo MPI biodistribution measurements of iron in liver, spleen, heart, and lungs after injection showed excellent agreement (R2 = 0.943) with measurements from induction coupled plasma spectrometry. These results demonstrate that MPI offers strong utility for noninvasively imaging and quantifying the systemic distribution of cell therapies and other therapeutic agents.
Collapse
|
39
|
Abstract
Lineage tracing is a widely used method for understanding cellular dynamics in multicellular organisms during processes such as development, adult tissue maintenance, injury repair and tumorigenesis. Advances in tracing or tracking methods, from light microscopy-based live cell tracking to fluorescent label-tracing with two-photon microscopy, together with emerging tissue clearing strategies and intravital imaging approaches have enabled scientists to decipher adult stem and progenitor cell properties in various tissues and in a wide variety of biological processes. Although technical advances have enabled time-controlled genetic labeling and simultaneous live imaging, a number of obstacles still need to be overcome. In this review, we aim to provide an in-depth description of the traditional use of lineage tracing as well as current strategies and upcoming new methods of labeling and imaging.
Collapse
Affiliation(s)
| | | | - Bon-Kyoung Koo
- Department of Genetics and Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, United Kingdom
| |
Collapse
|
40
|
He X, Cai J, Liu B, Zhong Y, Qin Y. Cellular magnetic resonance imaging contrast generated by the ferritin heavy chain genetic reporter under the control of a Tet-On switch. Stem Cell Res Ther 2015; 6:207. [PMID: 26517988 PMCID: PMC4628232 DOI: 10.1186/s13287-015-0205-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/30/2015] [Accepted: 10/16/2015] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Despite the strong appeal of ferritin as a magnetic resonance imaging (MRI) reporter for stem cell research, no attempts have been made to apply this genetic imaging reporter in stem cells in an inducible manner, which is important for minimizing the potential risk related to the constitutive expression of an imaging reporter. The aim of the present study was to develop an inducible genetic MRI reporter system that enables the production of intracellular MRI contrast as needed. METHODS Ferritin heavy chain (FTH1) was genetically modified by adding a Tet-On switch. A C3H10T1/2 cell line carrying Tet-FTH1 (C3H10T1/2-FTH1) was established via lentiviral transduction. The dose- and time-dependent expression of FTH1 in C3H10T1/2 cells was assessed by western blot and immunofluorescence staining. The induced "ON" and non-induced "OFF" expressions of FTH1 were detected using a 3.0 T MRI scanner. Iron accumulation in cells was analyzed by Prussian blue staining and transmission electron microscopy (TEM). RESULTS The expression of FTH1 was both dose- and time-dependently induced, and FTH1 expression peaked in response to induction with doxycycline (Dox) at 0.2 μg/ml for 72 h. The induced expression of FTH1 resulted in a significant increase in the transverse relaxation rate of C3H10T1/2-FTH1 cells following iron supplementation. Prussian blue staining and TEM revealed extensive iron accumulation in C3H10T1/2-FTH1 cells in the presence of Dox. CONCLUSIONS Cellular MRI contrast can be produced as needed via the expression of FTH1 under the control of a Tet-On switch. This finding could lay the groundwork for the use of FTH1 to track stem cells in vivo in an inducible manner.
Collapse
Affiliation(s)
- Xiaoya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Jinhua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Bo Liu
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Yi Zhong
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
41
|
Guan X, Jiang X, Yang C, Tian X, Li L. The MRI marker geneMagAattenuates the oxidative damage induced by iron overload in transgenic mice. Nanotoxicology 2015; 10:531-41. [DOI: 10.3109/17435390.2015.1090029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Bernsen MR, Guenoun J, van Tiel ST, Krestin GP. Nanoparticles and clinically applicable cell tracking. Br J Radiol 2015; 88:20150375. [PMID: 26248872 DOI: 10.1259/bjr.20150375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In vivo cell tracking has emerged as a much sought after tool for design and monitoring of cell-based treatment strategies. Various techniques are available for pre-clinical animal studies, from which much has been learned and still can be learned. However, there is also a need for clinically translatable techniques. Central to in vivo cell imaging is labelling of cells with agents that can give rise to signals in vivo, that can be detected and measured non-invasively. The current imaging technology of choice for clinical translation is MRI in combination with labelling of cells with magnetic agents. The main challenge encountered during the cell labelling procedure is to efficiently incorporate the label into the cell, such that the labelled cells can be imaged at high sensitivity for prolonged periods of time, without the labelling process affecting the functionality of the cells. In this respect, nanoparticles offer attractive features since their structure and chemical properties can be modified to facilitate cellular incorporation and because they can carry a high payload of the relevant label into cells. While these technologies have already been applied in clinical trials and have increased the understanding of cell-based therapy mechanism, many challenges are still faced.
Collapse
Affiliation(s)
- Monique R Bernsen
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands.,2 Department of Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jamal Guenoun
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands
| | | | | |
Collapse
|
43
|
Bakermans AJ, Abdurrachim D, Moonen RPM, Motaal AG, Prompers JJ, Strijkers GJ, Vandoorne K, Nicolay K. Small animal cardiovascular MR imaging and spectroscopy. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2015; 88-89:1-47. [PMID: 26282195 DOI: 10.1016/j.pnmrs.2015.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 06/04/2023]
Abstract
The use of MR imaging and spectroscopy for studying cardiovascular disease processes in small animals has increased tremendously over the past decade. This is the result of the remarkable advances in MR technologies and the increased availability of genetically modified mice. MR techniques provide a window on the entire timeline of cardiovascular disease development, ranging from subtle early changes in myocardial metabolism that often mark disease onset to severe myocardial dysfunction associated with end-stage heart failure. MR imaging and spectroscopy techniques play an important role in basic cardiovascular research and in cardiovascular disease diagnosis and therapy follow-up. This is due to the broad range of functional, structural and metabolic parameters that can be quantified by MR under in vivo conditions non-invasively. This review describes the spectrum of MR techniques that are employed in small animal cardiovascular disease research and how the technological challenges resulting from the small dimensions of heart and blood vessels as well as high heart and respiratory rates, particularly in mice, are tackled.
Collapse
Affiliation(s)
- Adrianus J Bakermans
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rik P M Moonen
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abdallah G Motaal
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Gustav J Strijkers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katrien Vandoorne
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
44
|
Overexpression of the MRI Reporter Genes Ferritin and Transferrin Receptor Affect Iron Homeostasis and Produce Limited Contrast in Mesenchymal Stem Cells. Int J Mol Sci 2015; 16:15481-96. [PMID: 26184159 PMCID: PMC4519909 DOI: 10.3390/ijms160715481] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/24/2015] [Accepted: 07/02/2015] [Indexed: 11/17/2022] Open
Abstract
Imaging technologies that allow the non-invasive monitoring of stem cells in vivo play a vital role in cell-based regenerative therapies. Recently, much interest has been generated in reporter genes that enable simultaneous monitoring of the anatomical location and viability of cells using magnetic resonance imaging (MRI). Here, we investigate the efficacy of ferritin heavy chain-1 (Fth1) and transferrin receptor-1 (TfR1) as reporters for tracking mesenchymal stem cells. The overexpression of TfR1 was well tolerated by the cells but Fth1 was found to affect the cell’s iron homeostasis, leading to phenotypic changes in the absence of iron supplementation and an upregulation in transcript and protein levels of the cell’s endogenous transferrin receptor. Neither the sole overexpression of Fth1 nor TfR1 resulted in significant increases in intracellular iron content, although significant differences were seen when the two reporter genes were used in combination, in the presence of high concentrations of iron. The supplementation of the culture medium with iron sources was a more efficient means to obtain contrast than the use of reporter genes, where high levels of intracellular iron were reflected in transverse (T2) relaxation. The feasibility of imaging iron-supplemented cells by MRI is shown using a 3R-compliant chick embryo model.
Collapse
|
45
|
Srivastava AK, Kadayakkara DK, Bar-Shir A, Gilad AA, McMahon MT, Bulte JWM. Advances in using MRI probes and sensors for in vivo cell tracking as applied to regenerative medicine. Dis Model Mech 2015; 8:323-36. [PMID: 26035841 PMCID: PMC4381332 DOI: 10.1242/dmm.018499] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The field of molecular and cellular imaging allows molecules and cells to be visualized in vivo non-invasively. It has uses not only as a research tool but in clinical settings as well, for example in monitoring cell-based regenerative therapies, in which cells are transplanted to replace degenerating or damaged tissues, or to restore a physiological function. The success of such cell-based therapies depends on several critical issues, including the route and accuracy of cell transplantation, the fate of cells after transplantation, and the interaction of engrafted cells with the host microenvironment. To assess these issues, it is necessary to monitor transplanted cells non-invasively in real-time. Magnetic resonance imaging (MRI) is a tool uniquely suited to this task, given its ability to image deep inside tissue with high temporal resolution and sensitivity. Extraordinary efforts have recently been made to improve cellular MRI as applied to regenerative medicine, by developing more advanced contrast agents for use as probes and sensors. These advances enable the non-invasive monitoring of cell fate and, more recently, that of the different cellular functions of living cells, such as their enzymatic activity and gene expression, as well as their time point of cell death. We present here a review of recent advancements in the development of these probes and sensors, and of their functioning, applications and limitations.
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, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Deepak K Kadayakkara
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amnon Bar-Shir
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Assaf A Gilad
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Michael T McMahon
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA. Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Öz G, Kittelson E, Demirgöz D, Rainwater O, Eberly LE, Orr HT, Clark HB. Assessing recovery from neurodegeneration in spinocerebellar ataxia 1: Comparison of in vivo magnetic resonance spectroscopy with motor testing, gene expression and histology. Neurobiol Dis 2015; 74:158-66. [PMID: 25446943 PMCID: PMC4323665 DOI: 10.1016/j.nbd.2014.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/18/2014] [Accepted: 11/19/2014] [Indexed: 12/29/2022] Open
Abstract
Suppression of transgene expression in a conditional transgenic mouse model of spinocerebellar ataxia 1 (SCA1) reverses the Purkinje cell pathology and motor dysfunction that are hallmarks of SCA1. We previously showed that cerebellar neurochemical levels measured by magnetic resonance spectroscopy (MRS) correlate with progression of pathology and clinical status of patients and that abnormal neurochemical levels normalize upon suppression of transgene expression, indicating their potential as robust surrogate markers of treatment effects. Here we investigated the relative sensitivities of MRS, histology, transgene expression and motor behavioral testing to disease reversal in conditional SCA1 mice. Transgene expression was suppressed by doxycycline administration and treated and untreated mice were assessed by MRS at 9.4tesla before and after treatment and with an accelerating Rotarod, histology and quantitative polymerase chain reaction (qPCR) for ataxin-1 transgene expression following doxycycline treatment. The MRS-measured N-acetylaspartate-to-myo-inositol ratio (NAA/Ins) correlated significantly with the molecular layer (ML) thickness and transgene expression. NAA/Ins, ML thickness and transgene expression were highly significantly different between the treated vs. untreated groups (p<0.0001), while the Rotarod assessment showed a trend for treatment effect. MRS, qPCR and histology had high sensitivity/specificity to distinguish treated from untreated mice, all with areas under the curve (AUC)=0.97-0.98 in receiver operating characteristic (ROC) analyses, while Rotarod had significantly lower sensitivity and specificity (AUC=0.72). Therefore, MRS accurately reflects the extent of recovery from neurodegeneration with sensitivity similar to invasive measures, further validating its potential as a surrogate marker in pre-clinical and clinical treatment trials.
Collapse
Affiliation(s)
- Gülin Öz
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, 2021 6th St. SE, Minneapolis, MN 55455, USA.
| | - Emily Kittelson
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, 2021 6th St. SE, Minneapolis, MN 55455, USA
| | - Döne Demirgöz
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, 2021 6th St. SE, Minneapolis, MN 55455, USA
| | - Orion Rainwater
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Lynn E Eberly
- Division of Biostatistics, School of Public Health, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - H Brent Clark
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Vande Velde G, Himmelreich U, Neeman M. Reporter gene approaches for mapping cell fate decisions by MRI: promises and pitfalls. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 8:424-31. [PMID: 24375898 DOI: 10.1002/cmmi.1590] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/29/2013] [Indexed: 12/15/2022]
Abstract
The central dogma of molecular biology, namely the process by which information encoded in the DNA serves as the template for transcriptional activation of specific mRNA resulting in temporal and spatial control of the translation of specific proteins, stands at the basis of normal and pathological cellular processes. Serving as the primary mechanism linking genotype to phenotype, it is clearly of significant interest for in vivo imaging. While classically, imaging revolutionized the ability to phenotype the anatomical and physiological impact of induction of changes in gene expression, the preceding molecular events remained invisible. Reporter gene-based imaging techniques provide a window for in vivo visualization of such transcriptional activation events. In addition to the widespread use of fluorescent and bioluminescent reporter genes and development of a number of reporter genes for positron emission tomography (PET) imaging, there has been significant progress in the development of reporter genes for MRI. With the development of strategies for cellular based therapies, such imaging tools could become central components for personalized patient monitoring.
Collapse
Affiliation(s)
- Greetje Vande Velde
- Biomedical MRI, KU Leuven, O&N I Herestraat 49 - box 505, 3000, Leuven, Belgium
| | | | | |
Collapse
|
48
|
Cho IK, Moran SP, Paudyal R, Piotrowska-Nitsche K, Cheng PH, Zhang X, Mao H, Chan AW. Longitudinal monitoring of stem cell grafts in vivo using magnetic resonance imaging with inducible maga as a genetic reporter. Am J Cancer Res 2014; 4:972-89. [PMID: 25161700 PMCID: PMC4143941 DOI: 10.7150/thno.9436] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/29/2014] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The ability to longitudinally monitor cell grafts and assess their condition is critical for the clinical translation of stem cell therapy in regenerative medicine. Developing an inducible genetic magnetic resonance imaging (MRI) reporter will enable non-invasive and longitudinal monitoring of stem cell grafts in vivo. METHODS MagA, a bacterial gene involved in the formation of iron oxide nanocrystals, was genetically modified for in vivo monitoring of cell grafts by MRI. Inducible expression of MagA was regulated by a Tet-On (Tet) switch. A mouse embryonic stem cell-line carrying Tet-MagA (mESC-MagA) was established by lentivirus transduction. The impact of expressing MagA in mESCs was evaluated via proliferation assay, cytotoxicity assay, teratoma formation, MRI, and inductively coupled plasma atomic emission spectroscopy (ICP-OES). Mice were grafted with mESCs with and without MagA (mESC-MagA and mESC-WT). The condition of cell grafts with induced "ON" and non-induced "OFF" expression of MagA was longitudinally monitored in vivo using a 7T MRI scanner. After imaging, whole brain samples were harvested for histological assessment. RESULTS Expression of MagA in mESCs resulted in significant changes in the transverse relaxation rate (R2 or 1/T2) and susceptibility weighted MRI contrast. The pluripotency of mESCs carrying MagA was not affected in vitro or in vivo. Intracranial mESC-MagA grafts generated sufficient T2 and susceptibility weighted contrast at 7T. The mESC-MagA grafts can be monitored by MRI longitudinally upon induced expression of MagA by administering doxycycline (Dox) via diet. CONCLUSION Our results demonstrate MagA could be used to monitor cell grafts noninvasively, longitudinally, and repetitively, enabling the assessment of cell graft conditions in vivo.
Collapse
|
49
|
Aryal S, Key J, Stigliano C, Landis MD, Lee DY, Decuzzi P. Positron emitting magnetic nanoconstructs for PET/MR imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:2688-2696. [PMID: 24639392 DOI: 10.1002/smll.201303933] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/10/2014] [Indexed: 06/03/2023]
Abstract
Hybrid PET/MRI scanners have the potential to provide fundamental molecular, cellular, and anatomic information essential for optimizing therapeutic and surgical interventions. However, their full utilization is currently limited by the lack of truly multi-modal contrast agents capable of exploiting the strengths of each modality. Here, we report on the development of long-circulating positron-emitting magnetic nanoconstructs (PEM) designed to image solid tumors for combined PET/MRI. PEMs are synthesized by a modified nano-precipitation method mixing poly(lactic-co-glycolic acid) (PLGA), lipids, and polyethylene glycol (PEG) chains with 5 nm iron oxide nanoparticles (USPIOs). PEM lipids are coupled with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and subsequently chelated to (64)Cu. PEMs show a diameter of 140 ± 7 nm and a transversal relaxivity r2 of 265.0 ± 10.0 (mM × s)(-1), with a r2/r1 ratio of 123. Using a murine xenograft model bearing human breast cancer cell line (MDA-MB-231), intravenously administered PEMs progressively accumulate in tumors reaching a maximum of 3.5 ± 0.25% ID/g tumor at 20 h post-injection. Correlation of PET and MRI signals revealed non-uniform intratumoral distribution of PEMs with focal areas of accumulation at the tumor periphery. These long-circulating PEMs with high transversal relaxivity and tumor accumulation may allow for detailed interrogation over multiple scales in a clinically relevant setting.
Collapse
Affiliation(s)
- Santosh Aryal
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Recent developments and improvements of multimodal imaging methods for use in animal research have substantially strengthened the options of in vivo visualization of cancer-related processes over time. Moreover, technological developments in probe synthesis and labelling have resulted in imaging probes with the potential for basic research, as well as for translational and clinical applications. In addition, more sophisticated cancer models are available to address cancer-related research questions. This Review gives an overview of developments in these three fields, with a focus on imaging approaches in animal cancer models and how these can help the translation of new therapies into the clinic.
Collapse
Affiliation(s)
- Marion de Jong
- Departments of Nuclear Medicine and Radiology, Erasmus MC Rotterdam, Room Na-610, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jeroen Essers
- Departments of Genetics (Cancer Genomics Centre), Radiation Oncology and Vascular Surgery, Erasmus MC Rotterdam, P.O Box 2040, 3000CA Rotterdam, The Netherlands
| | - Wytske M van Weerden
- Department of Urology, Erasmus MC Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|