1
|
McRae S, Martinez F, Foster P, Ronald J, Scholl T. Single-Frequency Birdcage Coils for Deep Tissue Perfluorocarbon Magnetic Resonance Imaging in Mice. NMR IN BIOMEDICINE 2025; 38:e5296. [PMID: 39648071 PMCID: PMC11625660 DOI: 10.1002/nbm.5296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 12/10/2024]
Abstract
Fluorine-19 (19F) MRI has become an established tool for in vivo cell tracking following ex vivo or in vivo labelling of various cell types with 19F perfluorocarbons (PFCs). Here, we developed and evaluated novel mouse-specific radiofrequency (RF) hardware for improved dual 1H anatomical imaging and deep tissue 19F MR detection of PFCs. Three linearly polarized birdcage RF coils were constructed-a dual-frequency 1H/19F coil, and a pair of single-frequency 1H and 19F coils, designed to be used sequentially. RF coil quality factors (Q values), signal homogeneity and sensitivity were benchmarked against a commercially constructed dual-frequency 1H/19F surface coil. RF homogeneity was assessed using a phantom designed to mimic PFC localization at depth in a mouse. The single-frequency birdcage coils (1H and 19F) displayed more uniform coverage and enhanced signal-to-noise ratios (SNRs) compared to both the birdcage and surface dual-frequency coils for 19F detection. Bilateral injection of a perfluoropolyether nanoemulsion into the footpads of female athymic nude mice, resulting in drainage to various lymph nodes and subsequent accumulation in lymph node macrophages, provided a platform to assess differences in SNRs and contrast-to-noise ratios (CNR) between both coil configurations as a function of depth and location. The single-frequency 1H coil provided significantly increased CNR in anatomical images (p < 0.001) with increased anatomical coverage compared to the dual-frequency surface coil. The single-frequency 19F birdcage coil offered increased PFC detectability with significantly higher SNR in renal, lumbar, sciatic and popliteal lymph nodes (p < 0.01) compared to the dual-frequency surface coil. Interestingly, the percentage difference between SNR measurements in lymph nodes between the single-frequency 19F coil and the 1H/19F surface coil had a linear relationship with increasing distance from the surface coil (R2 = 0.6352; p < 0.0001), indicating a potential disagreement for imaging experiments that rely on 19F spin quantification at increasing depth within the mouse using surface RF coils.
Collapse
Affiliation(s)
- Sean W. McRae
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
| | | | - Paula J. Foster
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's Health CareLondonOntarioCanada
| | - John A. Ronald
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's Health CareLondonOntarioCanada
| | - Timothy J. Scholl
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Department of Physics and AstronomyUniversity of Western OntarioLondonOntarioCanada
- Ontario Institute for Cancer ResearchTorontoOntarioCanada
| |
Collapse
|
2
|
Navarrete-León C, Doherty A, Strimaite M, Bear JC, Olivo A, Endrizzi M, Patrick PS. Nanoparticle Contrast Agents for Dark-Field X-ray Imaging. NANO LETTERS 2024. [PMID: 39601295 DOI: 10.1021/acs.nanolett.4c04878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The poor soft tissue contrast of X-ray CT necessitates contrast agent use to improve diagnosis across disease applications, yet their poor detection sensitivity requires high injected doses, which restrict use in at-risk populations. Dark-field X-ray imaging is emerging as a more sensitive alternative to traditional attenuation-based imaging, leveraging scattered radiation to produce contrast. Yet aside from large, short-lived microbubbles, the alternate physics of dark-field detection has yet to be exploited for contrast agent development. Here we demonstrate that high-Z nanoparticles can provide a new means to producing dark-field image contrast, promoting scatter via a higher rather than lower electron density compared to microbubbles, increasing detection sensitivity compared to attenuation-based detection of a clinical iodine-based agent at an equivalent X-ray dose. As the use of dark-field X-ray imaging expands into more common clinical usage, this will support the development of a new class of nanoparticulate contrast agents.
Collapse
Affiliation(s)
- Carlos Navarrete-León
- Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray microscopy and tomography lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Adam Doherty
- Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray microscopy and tomography lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Margarita Strimaite
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
- UCL School of Pharmacy, Faculty of Life Sciences, University College London, London, WC1N 1AX, United Kingdom
| | - Joseph C Bear
- School of Life Sciences, Pharmacy & Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, United Kingdom
| | - Alessandro Olivo
- Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
| | - Marco Endrizzi
- Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray microscopy and tomography lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - P Stephen Patrick
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| |
Collapse
|
3
|
Allouche-Arnon H, Montrazi ET, Subramani B, Fisler M, Spigel I, Frydman L, Mehlman T, Brandis A, Harris T, Bar-Shir A. A Genetically Engineered Reporter System Designed for 2H-MRI Allows Quantitative In Vivo Mapping of Transgene Expression. J Am Chem Soc 2024; 146:31624-31632. [PMID: 39527270 PMCID: PMC11583250 DOI: 10.1021/jacs.4c09572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The ability to obtain quantitative spatial information on subcellular processes of deep tissues in vivo has been a long-standing challenge for molecular magnetic resonance imaging (MRI) approaches. This challenge remains even more so for quantifying readouts of genetically engineered MRI reporters. Here, we set to overcome this challenge with a molecular system designed to obtain quantitative 2H-MRI maps of a gene reporter. To this end, we synthesized deuterated thymidine, d3-thy, with three magnetically equivalent deuterons at its methyl group (-CD3), showing a singlet peak with a characteristic 2H-NMR frequency (δ = 1.7 ppm). The upfield 3.0 ppm offset from the chemical shift of the HDO signal (δ = 4.7 ppm) allows for spectrally resolving the two 2H NMR signals and quantifying the concentration of d3-thy based on the known concentration of a tissue's HDO. Following systemic administration of d3-thy, its accumulation as d3-thy monophosphate in cells expressing the human thymidine kinase 1 (hTK1) transgene was mapped with 2H-MRI. The data obtained in vivo show the ability to use the d3-thy/hTK1 pair as a reporter probe/reporter gene system to quantitatively map transgene expression with MRI. Relying on a structurally unmodified reporter probe (d3-thy) to image the expression of unmutated human protein (hTK1) shows the potential of molecular imaging with 2H-MRI to monitor gene reporters and other relevant biological targets.
Collapse
Affiliation(s)
- Hyla Allouche-Arnon
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elton T. Montrazi
- Department
of Chemical and Biological Physics, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Balamurugan Subramani
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Fisler
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Inbal Spigel
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lucio Frydman
- Department
of Chemical and Biological Physics, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Tevie Mehlman
- Department
of Life Sciences Core Facilities, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Alexander Brandis
- Department
of Life Sciences Core Facilities, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Talia Harris
- Department
of Chemical Research Support, Weizmann Institute
of Science, Rehovot 7610001, Israel
| | - Amnon Bar-Shir
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
4
|
Ning Y, Yuwen Zhou I, Caravan P. Quantitative in Vivo Molecular MRI. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407262. [PMID: 39279542 PMCID: PMC11530320 DOI: 10.1002/adma.202407262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Molecular magnetic resonance imaging (MRI) combines chemistry, chemical biology, and imaging techniques to track molecular events non-invasively. Quantitative molecular MRI aims to provide meaningful, reproducible numerical measurements of molecular processes or biochemical targets within the body. In this review, the classifications of molecular MRI probes based on their signal-generating mechanism and functionality are first described. From there, the primary considerations for in vitro characterization and in vivo validation of molecular MRI probes, including how to avoid pitfalls and biases are discussed. Then, recommendations on imaging acquisition protocols and analysis methods to establish quantitative relationships between MRI signal change induced by the probes and the molecular processes of interest are provided. Finally, several representative case studies are highlighted that incorporate these features. Quantitative molecular MRI is a multidisciplinary research area incorporating expertise in chemical biology, inorganic chemistry, molecular probes, imaging physics, drug development, pathobiology, and medicine. The purpose of this review is to provide guidance to chemists developing MR imaging probes and methods in terms of in vitro and in vivo validation to accelerate the translation of these new quantitative tools for non-invasive imaging of biological processes.
Collapse
Affiliation(s)
- Yingying Ning
- Spin-X Institute, School of Chemistry and Chemical Engineering, School of Biomedical Sciences and Engineering, State Key Laboratory of Luminescent Materials and Devices, Guangdong-Hong Kong-Macao Joint Laboratory of Optoelectronic and Magnetic Functional Materials, South China University of Technology, Guangzhou 510641, China
| | - Iris Yuwen Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
5
|
Chacko AN, Miller ADC, Dhanabalan KM, Mukherjee A. Exploring the potential of water channels for developing genetically encoded reporters and biosensors for diffusion-weighted MRI. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2024; 365:107743. [PMID: 39053029 PMCID: PMC11687277 DOI: 10.1016/j.jmr.2024.107743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Genetically encoded reporters for magnetic resonance imaging (MRI) offer a valuable technology for making molecular-scale measurements of biological processes within living organisms with high anatomical resolution and whole-organ coverage without relying on ionizing radiation. However, most MRI reporters rely on synthetic contrast agents, typically paramagnetic metals and metal complexes, which often need to be supplemented exogenously to create optimal contrast. To eliminate the need for synthetic contrast agents, we previously introduced aquaporin-1, a mammalian water channel, as a new reporter gene for the fully autonomous detection of genetically labeled cells using diffusion-weighted MRI. In this study, we aimed to expand the toolbox of diffusion-based genetic reporters by modulating aquaporin membrane trafficking and harnessing the evolutionary diversity of water channels across species. We identified a number of new water channels that functioned as diffusion-weighted reporter genes. In addition, we show that loss-of-function variants of yeast and human aquaporins can be leveraged to design first-in-class diffusion-based sensors for detecting the activity of a model protease within living cells.
Collapse
Affiliation(s)
- Asish N Chacko
- Department of Chemistry, University of California, Santa Barbara, CA 93106-5080, USA
| | - Austin D C Miller
- Biomolecular Science and Engineering Graduate Program, University of California, Santa Barbara, CA 93106-5080, USA
| | - Kaamini M Dhanabalan
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106-5080, USA
| | - Arnab Mukherjee
- Department of Chemistry, University of California, Santa Barbara, CA 93106-5080, USA; Biomolecular Science and Engineering Graduate Program, University of California, Santa Barbara, CA 93106-5080, USA; Department of Chemical Engineering, University of California, Santa Barbara, CA 93106-5080, USA; Department of Bioengineering, University of California, Santa Barbara, CA 93106-5080, USA.
| |
Collapse
|
6
|
Rallapalli H, McCall EC, Koretsky AP. Genetic control of MRI contrast using the manganese transporter Zip14. Magn Reson Med 2024; 92:820-835. [PMID: 38573932 PMCID: PMC11142883 DOI: 10.1002/mrm.29993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/15/2023] [Accepted: 12/12/2023] [Indexed: 04/06/2024]
Abstract
PURPOSE Gene-expression reporter systems, such as green fluorescent protein, have been instrumental to understanding biological processes in living organisms at organ system, tissue, cell, and molecular scales. More than 30 years of work on developing MRI-visible gene-expression reporter systems has resulted in a variety of clever application-specific methods. However, these techniques have not yet been widely adopted, so a general-purpose expression reporter is still required. Here, we demonstrate that the manganese ion transporter Zip14 is an in vivo MRI-visible, flexible, and robust gene-expression reporter to meet this need. METHODS Plasmid constructs consisting of a cell type-specific promoter, gene coding for human Zip14, and a histology-visible tag were packaged into adeno-associated viruses. These viruses were intracranially injected into the mouse brain. Serial in vivo MRI was performed using a vendor-supplied 3D-MPRAGE sequence. No additional contrast agents were administered. Animals were sacrificed after the last imaging timepoint for immunohistological validation. RESULTS Neuron-specific overexpression of Zip14 produced substantial and long-lasting changes in MRI contrast. Using appropriate viruses enabled both anterograde and retrograde neural tracing. Expression of Zip14 in astrocytes also enabled MRI of glia populations in the living mammalian brain. CONCLUSIONS The flexibility of this system as an MRI-visible gene-expression reporter will enable many applications of serial, high-resolution imaging of gene expression for basic science and therapy development.
Collapse
Affiliation(s)
- Harikrishna Rallapalli
- Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
| | - Eleanor C McCall
- Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
| | - Alan P Koretsky
- Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Bettini A, Patrick PS, Day RM, Stuckey DJ. CT-Visible Microspheres Enable Whole-Body In Vivo Tracking of Injectable Tissue Engineering Scaffolds. Adv Healthc Mater 2024; 13:e2303588. [PMID: 38678393 PMCID: PMC11468734 DOI: 10.1002/adhm.202303588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/27/2024] [Indexed: 04/30/2024]
Abstract
Targeted delivery and retention are essential requirements for implantable tissue-engineered products. Non-invasive imaging methods that can confirm location, retention, and biodistribution of transplanted cells attached to implanted tissue engineering scaffolds will be invaluable for the optimization and enhancement of regenerative therapies. To address this need, an injectable tissue engineering scaffold consisting of highly porous microspheres compatible with transplantation of cells is modified to contain the computed tomography (CT) contrast agent barium sulphate (BaSO4). The trackable microspheres show high x-ray absorption, with contrast permitting whole-body tracking. The microspheres are cellularized with GFP+ Luciferase+ mesenchymal stem cells and show in vitro biocompatibility. In vivo, cellularized BaSO4-loaded microspheres are delivered into the hindlimb of mice where they remain viable for 14 days. Co-registration of 3D-bioluminescent imaging and µCT reconstructions enable the assessment of scaffold material and cell co-localization. The trackable microspheres are also compatible with minimally-invasive delivery by ultrasound-guided transthoracic intramyocardial injections in rats. These findings suggest that BaSO4-loaded microspheres can be used as a novel tool for optimizing delivery techniques and tracking persistence and distribution of implanted scaffold materials. Additionally, the microspheres can be cellularized and have the potential to be developed into an injectable tissue-engineered combination product for cardiac regeneration.
Collapse
Affiliation(s)
- Annalisa Bettini
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College LondonLondonWC1E 6DDUK
- Centre for Precision HealthcareDivision of MedicineUniversity College LondonLondonWC1E 6JFUK
| | - Peter Stephen Patrick
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College LondonLondonWC1E 6DDUK
| | - Richard M. Day
- Centre for Precision HealthcareDivision of MedicineUniversity College LondonLondonWC1E 6JFUK
| | - Daniel J. Stuckey
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College LondonLondonWC1E 6DDUK
| |
Collapse
|
8
|
Iqbal Z, Azad R, Chen XS, Lin XL, Zhou Z, Wang XM, Nie RE. A New Species of Scymnus (Coleoptera, Coccinellidae) from Pakistan with Mitochondrial Genome and Its Phylogenetic Implications. INSECTS 2024; 15:371. [PMID: 38786927 PMCID: PMC11122443 DOI: 10.3390/insects15050371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
In this study, a new species of the subgenus Pullus belonging to the Scymnus genus from Pakistan, Scymnus (Pullus) cardi sp. nov., was described and illustrated, with information on its distribution, host plants, and prey. Additionally, the completed mitochondrial genome (mitogenome) of the new species using high-throughput sequencing technology was obtained. The genome contains the typical 37 genes (13 protein-coding genes, two ribosomal RNAs, and 22 transfer RNAs) and a non-coding control region, and is arranged in the same order as that of the putative ancestor of beetles. The AT content of the mitogenome is approximately 85.1%, with AT skew and GC skew of 0.05 and -0.43, respectively. The calculated values of relative synonymous codon usage (RSCU) determine that the codon UUA (L) has the highest frequency. Furthermore, we explored the phylogenetic relationship among 59 representatives of the Coccinellidae using Bayesian inference and maximum likelihood methods, the results of which strongly support the monophyly of Coccinellinae. The phylogenetic results positioned Scymnus (Pullus) cardi in a well-supported clade with Scymnus (Pullus) loewii and Scymnus (Pullus) rubricaudus within the genus Scymnus and the tribe Scymnini. The mitochondrial sequence of S. (P.) cardi will contribute to the mitochondrial genome database and provide helpful information for the identification and phylogeny of Coccinellidae.
Collapse
Affiliation(s)
- Zafar Iqbal
- Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (Z.I.); (X.-L.L.)
| | - Rashid Azad
- Department of Entomology, The University of Haripur, Haripur 22620, Pakistan;
- Department of Entomology, South China Agricultural University, Guangzhou 510640, China
| | - Xiao-Sheng Chen
- Engineering Research Center of Biological Control, Ministry of Education, Guangzhou 510642, China;
- Department of Forest Protection, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Ling Lin
- Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (Z.I.); (X.-L.L.)
| | - Zichen Zhou
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2BX, UK;
- Department of Life Sciences, Natural History Museum, Cromwell Road, London SW7 5BD, UK
| | - Xing-Min Wang
- Department of Entomology, South China Agricultural University, Guangzhou 510640, China
- Engineering Technology Research Center of Agricultural Pest Biocontrol of Guangdong Province, Guangzhou 510640, China
| | - Rui-E Nie
- Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (Z.I.); (X.-L.L.)
| |
Collapse
|
9
|
Huang Y, Chen X, Zhu Z, Mukherjee A. A Dual-Gene Reporter-Amplifier Architecture for Enhancing the Sensitivity of Molecular MRI by Water Exchange. Chembiochem 2024; 25:e202400087. [PMID: 38439618 PMCID: PMC11604348 DOI: 10.1002/cbic.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/06/2024]
Abstract
The development of genetic reporters for magnetic resonance imaging (MRI) is essential for investigating biological functions in vivo. However, current MRI reporters have low sensitivity, making it challenging to create significant contrast against the tissue background, especially when only a small fraction of cells express the reporter. To overcome this limitation, we developed an approach for amplifying the sensitivity of molecular MRI by combining a chemogenetic contrast mechanism with a biophysical approach to increase water diffusion through the co-expression of a dual-gene construct comprising an organic anion transporting polypeptide, Oatp1b3, and a water channel, Aqp1. We first show that the expression of Aqp1 amplifies MRI contrast in cultured cells engineered to express Oatp1b3. We demonstrate that the contrast amplification is caused by Aqp1-driven increase in water exchange, which provides the gadolinium ions internalized by Oatp1b3-expressing cells with access to a larger water pool compared with exchange-limited conditions. We further show that our methodology allows cells to be detected using approximately 10-fold lower concentrations of gadolinium than that in the Aqp1-free scenario. Finally, we show that our approach enables the imaging of mixed-cell cultures containing a low fraction of Oatp1b3-labeled cells that are undetectable on the basis of Oatp1b3 expression alone.
Collapse
Affiliation(s)
| | - Xinyue Chen
- Department of Molecular, Cellular, and Developmental Biology
| | - Ziyue Zhu
- Department of Molecular, Cellular, and Developmental Biology
| | - Arnab Mukherjee
- Department of Chemistry
- Department of Molecular, Cellular, and Developmental Biology
- Department of Chemical Engineering
| |
Collapse
|
10
|
Bhattacharyya T, Mallett CL, Shapiro EM. MRI-Based Cell Tracking of OATP-Expressing Cell Transplants by Pre-Labeling with Gd-EOB-DTPA. Mol Imaging Biol 2024; 26:233-239. [PMID: 38448775 DOI: 10.1007/s11307-024-01904-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE A critical step in cell-based therapies is determining the exact position of transplanted cells immediately post-transplant. Here, we devised a method to detect cell transplants immediately post-transplant, using a clinical gadolinium-based contrast agent. These cells were detected as hyperintense signals using a clinically familiar T1-weighted MRI protocol. PROCEDURES HEK293 cells were stably transduced to express human OATP1B3, a hepatic organic anion transporting polypeptide that transports Gd-EOB-DTPA into cells that express the transporters, the intracellular accumulation of which cells causes signal enhancement on T1-weighted MRI. Cells were pre-labeled prior to injection in media containing Gd-EOB-DTPA for MRI evaluation and indocyanine green for cryofluorescence tomography validation. Labeled cells were injected into chicken hearts, in vitro, after which MRI and cryofluorescence tomography were performed in sequence. RESULTS OATP1B3-expressing cells had substantially reduced T1 following labeling with Gd-EOB-DTPA in culture. Following their implantation into chicken heart, these cells were robustly identified in T1-weighted MRI, with image-derived injection volumes of cells commensurate with intended injection volumes. Cryofluorescence tomography showed that the areas of signal enhancement in MRI overlapped with areas of indocyanine green signal, indicating that MRI signal enhancement was due to the transplanted cells. CONCLUSIONS OATP1B3-expressing cells can be pre-labeled with Gd-EOB-DTPA prior to injection into tissue, affording the use of clinically familiar T1-weighted MRI to robustly detect cell transplants immediately after transplant. This procedure is easily generalizable and has potential advantages over the use of iron oxide based cell labeling agents and imaging procedures.
Collapse
Affiliation(s)
- Tapas Bhattacharyya
- Molecular and Cellular Imaging Laboratory, Department of Radiology, Michigan State University, 846 Service Rd, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Christiane L Mallett
- Molecular and Cellular Imaging Laboratory, Department of Radiology, Michigan State University, 846 Service Rd, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Erik M Shapiro
- Molecular and Cellular Imaging Laboratory, Department of Radiology, Michigan State University, 846 Service Rd, East Lansing, MI, 48824, USA.
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
- Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
11
|
Huang Y, Chen X, Zhu Z, Mukherjee A. A dual-gene reporter-amplifier architecture for enhancing the sensitivity of molecular MRI by water exchange. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576672. [PMID: 38328134 PMCID: PMC10849537 DOI: 10.1101/2024.01.22.576672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The development of genetic reporters for magnetic resonance imaging (MRI) is essential for investigating biological functions in intact animals. However, current MRI reporters have low sensitivity, making it challenging to create significant contrast against the tissue background, especially when only a small percentage of cells express the reporter. To overcome this limitation, we developed an approach that amplifies signals by co-expressing an MRI reporter gene, Oatp1b3, with a water channel, aquaporin-1 (Aqp1). We first show that the expression of Aqp1 amplifies the paramagnetic relaxation effect of Oatp1b3 by facilitating transmembrane water exchange. This mechanism provides Oatp1b3-expressing cells with access to a larger water pool compared with typical exchange-limited conditions. We further demonstrated that our methodology allows dual-labeled cells to be detected using approximately 10-fold lower concentrations of contrast agent than that in the Aqp1-free scenario. Finally, we show that our approach enables the imaging of mixed-cell populations containing a low fraction of Oatp1b3-labeled cells that are otherwise undetectable based on Oatp1b3 expression alone.
Collapse
Affiliation(s)
| | - Xinyue Chen
- Department of Molecular, Cellular, and Developmental Biology
| | - Ziyue Zhu
- Department of Molecular, Cellular, and Developmental Biology
| | - Arnab Mukherjee
- Department of Chemistry
- Department of Molecular, Cellular, and Developmental Biology
| |
Collapse
|
12
|
Chacko AN, Miller AD, Dhanabalan KM, Mukherjee A. Exploring the potential of water channels for developing MRI reporters and sensors without the need for exogenous contrast agents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576580. [PMID: 38328035 PMCID: PMC10849501 DOI: 10.1101/2024.01.21.576580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Genetically encoded reporters for magnetic resonance imaging (MRI) offer a valuable technology for making molecular-scale measurements of biological processes within living organisms with high anatomical resolution and whole-organ coverage without relying on ionizing radiation. However, most MRI reporters rely on contrast agents, typically paramagnetic metals and metal complexes, which often need to be supplemented exogenously to create optimal contrast. To eliminate the need for contrast agents, we previously introduced aquaporin-1, a mammalian water channel, as a new reporter gene for the fully autonomous detection of genetically labeled cells using diffusion-weighted MRI. In this study, we aimed to expand the toolbox of diffusion-based genetic reporters by modulating aquaporin membrane trafficking and harnessing the evolutionary diversity of water channels across species. We identified a number of new water channels that functioned as diffusion-weighted reporter genes. In addition, we show that loss-of-function variants of yeast and human aquaporins can be leveraged to design first-in-class diffusion-based sensors for detecting the activity of a model protease within living cells.
Collapse
Affiliation(s)
| | | | | | - Arnab Mukherjee
- Department of Chemistry
- Biomolecular Science and Engineering Graduate Program
- Department of Chemical Engineering
| |
Collapse
|
13
|
Gao RR, Lei QL, Jin X, Zafar I, Yang XK, Su CY, Hao JS, Nie RE. Characterization of Four Complete Mitogenomes of Monolepta Species and Their Related Phylogenetic Implications. INSECTS 2024; 15:50. [PMID: 38249056 PMCID: PMC10816406 DOI: 10.3390/insects15010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
Monolepta is one of the diverse genera in the subfamily Galerucinae, including 708 species and 6 sub-species worldwide. To explore the information on the mitogenome characteristics and phylogeny of the section "Monoleptites", especially the genus Monolepta, we obtained the newly completed mitochondrial genomes (mitogenomes) of four Monolepta species using high-throughput sequencing technology. The lengths of these four new mitochondrial genomes are 16,672 bp, 16,965 bp, 16,012 bp, and 15,866 bp in size, respectively. All four mitochondrial genomes include 22 transfer RNA genes (tRNAs), 13 protein-coding genes (PCGs), 2 ribosomal RNA genes (rRNAs), and one control region, which is consistent with other Coleoptera. The results of the nonsynonymous with synonymous substitution rates showed that ND6 had the highest evolution rate, while COI displayed the lowest evolution rate. The substitution saturation of three datasets (13 PCGs_codon1, 13 PCGs_codon2, 13 PCGs_codon3) showed that there was no saturation across all datasets. Phylogenetic analyses based on three datasets (ND1, 15 genes of mitogenomes, and 13 PCGs_AA) were carried out using maximum likelihood (ML) and Bayesian inference (BI) methods. The results showed that mitogenomes had a greater capacity to resolve the main clades than the ND1 gene at the suprageneric and species levels. The section "Monoleptites" was proven to be a monophyletic group, while Monolepta was a non-monophyletic group. Based on ND1 data, the newly sequenced species whose antennal segment 2 was shorter than 3 were split into several clades, while, based on the mitogenomic dataset, the four newly sequenced species had close relationships with Paleosepharia. The species whose antennal segment 2 was as long as 3 were split into two clades, which indicated that the characteristic of "antennal segment 2 as long as 3" of the true "Monolepta" evolved multiple times in several subgroups. Therefore, to explore the relationships among the true Monolepta, the most important thing is to perform a thorough revision of Monolepta and related genera in the future.
Collapse
Affiliation(s)
- Rong-Rong Gao
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| | - Qi-Long Lei
- Department of Entomology, China Agricultural University, Beijing 100193, China;
| | - Xu Jin
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| | - Iqbal Zafar
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| | - Xing-Ke Yang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangdong Institute of Applied Biological Resources, Guangzhou 510260, China
| | - Cheng-Yong Su
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| | - Jia-Sheng Hao
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| | - Rui-E Nie
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (R.-R.G.); (X.J.); (I.Z.); (C.-Y.S.)
| |
Collapse
|
14
|
Bhattacharyya T, Mallett C, Shapiro EM. MRI-based cell tracking of OATP-expressing cell transplants by pre-labeling with Gd-EOB-DTPA. RESEARCH SQUARE 2023:rs.3.rs-3698429. [PMID: 38168297 PMCID: PMC10760244 DOI: 10.21203/rs.3.rs-3698429/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Purpose A critical step in cell-based therapies is determining the exact position of transplanted cells immediately post-transplant. Here, we devised a method to detect cell transplants immediately post-transplant, using a clinical gadolinium-based contrast agent. These cells were detected as hyperintense signals using a clinically familiar T1-weighted MRI protocol. Procedures HEK293 cells were stably transduced to express human OATP1B3, a hepatic organic anion transporting polypeptide that transports Gd-EOB-DTPA into cells that express the transporters, the intracellular accumulation of which cells causes signal enhancement on T1-weighted MRI. Cells were pre-labeled prior to injection in media containing Gd-EOB-DTPA for MRI evaluation and indocyanine green for cryofluorescence tomography validation. Labeled cells were injected into chicken hearts, in vitro, after which MRI and cryofluorescence tomography were performed in sequence. Results OATP1B3-expressing cells had substantially reduced T1 following labeling with Gd-EOB-DTPA in culture. Following their implantation into chicken heart, these cells were robustly identified in T1-weighted MRI, with image-derived injection volumes of cells commensurate with intended injection volumes. Cryofluorescence tomography showed that the areas of signal enhancement in MRI overlapped with areas of indocyanine green signal, indicating that MRI signal enhancement was due to the transplanted cells. Conclusions OATP1B3-expressing cells can be pre-labeled with Gd-EOB-DTPA prior to injection into tissue, affording the use of clinically familiar T1-weighted MRI to robustly detect cell transplants immediately after transplant. This procedure is easily generalizable and has potential advantages over the use of iron oxide based cell labeling agents and imaging procedures.
Collapse
|
15
|
Fillion AJ, Bricco AR, Lee HD, Korenchan D, Farrar CT, Gilad AA. Development of a synthetic biosensor for chemical exchange MRI utilizing in silico optimized peptides. NMR IN BIOMEDICINE 2023; 36:e5007. [PMID: 37469121 PMCID: PMC11075521 DOI: 10.1002/nbm.5007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI has been identified as a novel alternative to classical diagnostic imaging. Over the last several decades, many studies have been conducted to determine possible CEST agents, such as endogenously expressed compounds or proteins, that can be utilized to produce contrast with minimally invasive procedures and reduced or non-existent levels of toxicity. In recent years there has been an increased interest in the generation of genetically engineered CEST contrast agents, typically based on existing proteins with CEST contrast or modified to produce CEST contrast. We have developed an in silico method for the evolution of peptide sequences to optimize CEST contrast and showed that these peptides could be combined to create de novo biosensors for CEST MRI. A single protein, superCESTide, was designed to be 198 amino acids. SuperCESTide was expressed in E. coli and purified with size exclusion chromatography. The magnetic transfer ratio asymmetry generated by superCESTide was comparable to levels seen in previous CEST reporters, such as protamine sulfate (salmon protamine) and human protamine. These data show that novel peptides with sequences optimized in silico for CEST contrast that utilize a more comprehensive range of amino acids can still produce contrast when assembled into protein units expressed in complex living environments.
Collapse
Affiliation(s)
- Adam J. Fillion
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Harvey D. Lee
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - David Korenchan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Assaf A. Gilad
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
16
|
Gilad AA, Bar-Shir A, Bricco AR, Mohanta Z, McMahon MT. Protein and peptide engineering for chemical exchange saturation transfer imaging in the age of synthetic biology. NMR IN BIOMEDICINE 2023; 36:e4712. [PMID: 35150021 PMCID: PMC10642350 DOI: 10.1002/nbm.4712] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 05/23/2023]
Abstract
At the beginning of the millennium, the first chemical exchange saturation transfer (CEST) contrast agents were bio-organic molecules. However, later, metal-based CEST agents (paraCEST agents) took center stage. This did not last too long as paraCEST agents showed limited translational potential. By contrast, the CEST field gradually became dominated by metal-free CEST agents. One branch of research stemming from the original work by van Zijl and colleagues is the development of CEST agents based on polypeptides. Indeed, in the last 2 decades, tremendous progress has been achieved in this field. This includes the design of novel peptides as biosensors, genetically encoded recombinant as well as synthetic reporters. This was a result of extensive characterization and elucidation of the theoretical requirements for rational designing and engineering of such agents. Here, we provide an extensive overview of the evolution of more precise protein-based CEST agents, review the rationalization of enzyme-substrate pairs as CEST contrast enhancers, discuss the theoretical considerations to improve peptide selectivity, specificity and enhance CEST contrast. Moreover, we discuss the strong influence of synthetic biology on the development of the next generation of protein-based CEST contrast agents.
Collapse
Affiliation(s)
- Assaf A. Gilad
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Amnon Bar-Shir
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Zinia Mohanta
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Michael T. McMahon
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
17
|
McRae SW, Cleary M, DeRoche D, Martinez FM, Xia Y, Caravan P, Gale EM, Ronald JA, Scholl TJ. Development of a Suite of Gadolinium-Free OATP1-Targeted Paramagnetic Probes for Liver MRI. J Med Chem 2023; 66:6567-6576. [PMID: 37159947 DOI: 10.1021/acs.jmedchem.2c01561] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Five amphiphilic, anionic Mn(II) complexes were synthesized as contrast agents targeted to organic anion transporting polypeptide transporters (OATP) for liver magnetic resonance imaging (MRI). The Mn(II) complexes are synthesized in three steps, each from the commercially available trans-1,2-diaminocyclohexane-N,N,N',N'-tetraacetic acid (CDTA) chelator, with T1-relaxivity of complexes ranging between 2.3 and 3.0 mM-1 s-1 in phosphate buffered saline at an applied field strength of 3.0 T. Pharmacokinetics were assessed in female BALB/c mice by acquiring T1-weighted images dynamically for 70 min after agent administration and determining contrast enhancement and washout in various organs. Uptake of Mn(II) complexes in human OATPs was investigated through in vitro assays using MDA-MB-231 cells engineered to express either OATP1B1 or OATP1B3 isoforms. Our study introduces a new class of Mn-based OATP-targeted contrast that can be broadly tuned via simple synthetic protocols.
Collapse
Affiliation(s)
- Sean W McRae
- Department of Medical Biophysics, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Michael Cleary
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Daniel DeRoche
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Francisco M Martinez
- Imaging Laboratories, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Ying Xia
- Imaging Laboratories, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Peter Caravan
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Eric M Gale
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - John A Ronald
- Department of Medical Biophysics, University of Western Ontario, London, Ontario N6A 3K7, Canada
- Imaging Laboratories, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 3K7, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, University of Western Ontario, London, Ontario N6A 3K7, Canada
- Imaging Laboratories, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 3K7, Canada
- Department of Physics and Astronomy, University of Western Ontario, London, Ontario N6A 3K7, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| |
Collapse
|
18
|
Fillion AJ, Bricco AR, Lee HD, Korenchan D, Farrar CT, Gilad AA. Development of a Synthetic Biosensor for Chemical Exchange MRI Utilizing In Silico Optimized Peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531737. [PMID: 37016672 PMCID: PMC10071792 DOI: 10.1101/2023.03.08.531737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Chemical Exchange Saturation Transfer (CEST) magnetic resonance imaging (MRI) has been identified as a novel alternative to classical diagnostic imaging. Over the last several decades, many studies have been conducted to determine possible CEST agents, such as endogenously expressed compounds or proteins, that can be utilized to produce contrast with minimally invasive procedures and reduced or non-existent levels of toxicity. In recent years there has been an increased interest in the generation of genetically engineered CEST contrast agents, typically based on existing proteins with CEST contrast or modified to produce CEST contrast. We have developed an in-silico method for the evolution of peptide sequences to optimize CEST contrast and showed that these peptides could be combined to create de novo biosensors for CEST MRI. A single protein, superCESTide 2.0, was designed to be 198 amino acids. SuperCESTide 2.0 was expressed in E. coli and purified with size-exclusion chromatography. The magnetic transfer ratio asymmetry (MTR asym ) generated by superCESTide 2.0 was comparable to levels seen in previous CEST reporters, such as protamine sulfate (salmon protamine, SP), Poly-L-Lysine (PLL), and human protamine (hPRM1). This data shows that novel peptides with sequences optimized in silico for CEST contrast that utilizes a more comprehensive range of amino acids can still produce contrast when assembled into protein units expressed in complex living environments.
Collapse
Affiliation(s)
- Adam J. Fillion
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Harvey D. Lee
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - David Korenchan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Assaf A. Gilad
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
19
|
Nyström NN, McRae SW, Martinez FM, Kelly JJ, Scholl TJ, Ronald JA. A Genetically Encoded Magnetic Resonance Imaging Reporter Enables Sensitive Detection and Tracking of Spontaneous Metastases in Deep Tissues. Cancer Res 2023; 83:673-685. [PMID: 36512633 DOI: 10.1158/0008-5472.can-22-2770] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/11/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Metastasis is the leading cause of cancer-related death. However, it remains a poorly understood aspect of cancer biology, and most preclinical cancer studies do not examine metastasis, focusing solely on the primary tumor. One major factor contributing to this paradox is a gap in available tools for accurate spatiotemporal measurements of metastatic spread in vivo. Here, our objective was to develop an imaging reporter system that offers sensitive three-dimensional (3D) detection of cancer cells at high resolutions in live mice. An organic anion-transporting polypeptide 1b3 (oatp1b3) was used as an MRI reporter gene, and its sensitivity was systematically optimized for in vivo tracking of viable cancer cells in a spontaneous metastasis model. Metastases with oatp1b3-MRI could be observed at the single lymph node level and tracked over time as cancer cells spread to multiple lymph nodes and different organ systems in individual animals. While initial single lesions were successfully imaged in parallel via bioluminescence, later metastases were largely obscured by light scatter from the initial node. Importantly, MRI could detect micrometastases in lung tissue comprised on the order of 1,000 cancer cells. In summary, oatp1b3-MRI enables longitudinal tracking of cancer cells with combined high resolution and high sensitivity that provides 3D spatial information and the surrounding anatomical context. SIGNIFICANCE An MRI reporter gene system optimized for tracking metastasis in deep tissues at high resolutions and able to detect spontaneous micrometastases in lungs of mice provides a useful tool for metastasis research.
Collapse
Affiliation(s)
- Nivin N Nyström
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Department of Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Sean W McRae
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
| | - Francisco M Martinez
- Imaging Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
| | - John J Kelly
- Imaging Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada.,Department of Physics and Astronomy, Western University, London, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John A Ronald
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
20
|
Yang H, Qian Z, Liu C, Tie C, Cai A, Wang J, Xing Y, Xia J, Li X. A versatile genetic-encoded reporter for magnetic resonance imaging. Heliyon 2023; 9:e14054. [PMID: 36915487 PMCID: PMC10006841 DOI: 10.1016/j.heliyon.2023.e14054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
It has been a long-cherished wish in biomedicine research to have an imaging tool to visualize gene expression, with good spatiotemporal resolution, in rodent and primate animals noninvasively and longitudinally. To this purpose, we here present a novel genetic encoded magnetic resonance imaging reporter, i.e., GEM reporter, for noninvasive visualization of cell-specific gene expression. The GEM reporter was developed through codon modification of a bacteria-originated manganese (Mn) binding protein, allowing the sequestration of endogenous Mn in local tissues. When expressed in bacteria, plant and animals, GEM reporter can robustly produce high image contrast in T1-weighted MRI without additional substrates or contrast agents. Importantly, GEM reporter can be tracked inherently by MRI in specific cells and tissues. These findings support GEM reporter as a versatile marker for deciphering gene expression spatiotemporally in living subjects.
Collapse
Affiliation(s)
- Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhengjiang Qian
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chunhua Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Changjun Tie
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,Peng Cheng Laboratory, Shenzhen, China
| | - Aoling Cai
- University of Chinese Academy of Sciences, Beijing, 100049, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, 430071, Wuhan, Hubei Province, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, 430071, Wuhan, Hubei Province, China
| | - Yao Xing
- Shanghai United Imaging Healthcare Co., Ltd., China
| | - Jun Xia
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGang Road West, Shenzhen 518035, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
21
|
Cohen D, Allouche‐Arnon H, Bar‐Shir A. MRI reporter genes in the era of gene transfer. Clin Transl Med 2022; 12:e1135. [PMID: 36471476 PMCID: PMC9722966 DOI: 10.1002/ctm2.1135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Dan Cohen
- Department of Nuclear MedicineTel‐Aviv Sourasky Medical CenterTel AvivIsrael
| | - Hyla Allouche‐Arnon
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceRehovotIsrael
| | - Amnon Bar‐Shir
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
22
|
Hill M, Cunniffe N, Franklin R. Seeing is believing: Identifying remyelination in the central nervous system. Curr Opin Pharmacol 2022; 66:102269. [DOI: 10.1016/j.coph.2022.102269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
|
23
|
Brindle KM. Gene reporters for magnetic resonance imaging. Trends Genet 2022; 38:996-998. [PMID: 35641343 DOI: 10.1016/j.tig.2022.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022]
Abstract
MRI-based gene reporters allow imaging of gene expression at depth (tens of centimetres) and at relatively high resolution (~10-100 μm) and have the potential to be translated to the clinic. The reporters exploit either endogenous contrast mechanisms or they modulate the response to an introduced exogenous contrast agent.
Collapse
Affiliation(s)
- Kevin M Brindle
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, UK.
| |
Collapse
|
24
|
Abstract
MRI is a widely available clinical tool for cancer diagnosis and treatment monitoring. MRI provides excellent soft tissue imaging, using a wide range of contrast mechanisms, and can non-invasively detect tissue metabolites. These approaches can be used to distinguish cancer from normal tissues, to stratify tumor aggressiveness, and to identify changes within both the tumor and its microenvironment in response to therapy. In this review, the role of MRI in immunotherapy monitoring will be discussed and how it could be utilized in the future to address some of the unique clinical questions that arise from immunotherapy. For example, MRI could play a role in identifying pseudoprogression, mixed response, T cell infiltration, cell tracking, and some of the characteristic immune-related adverse events associated with these agents. The factors to be considered when developing MRI imaging biomarkers for immunotherapy will be reviewed. Finally, the advantages and limitations of each approach will be discussed, as well as the challenges for future clinical translation into routine clinical care. Given the increasing use of immunotherapy in a wide range of cancers and the ability of MRI to detect the microstructural and functional changes associated with successful response to immunotherapy, the technique has great potential for more widespread and routine use in the future for these applications.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Pippa G Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
25
|
Yun J, Baldini M, Chowdhury R, Mukherjee A. Designing Protein-Based Probes for Sensing Biological Analytes with Magnetic Resonance Imaging. ANALYSIS & SENSING 2022; 2:e202200019. [PMID: 37409177 PMCID: PMC10321474 DOI: 10.1002/anse.202200019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Genetically encoded sensors provide unique advantages for monitoring biological analytes with molecular and cellular-level specificity. While sensors derived from fluorescent proteins represent staple tools in biological imaging, these probes are limited to optically accessible preparations owing to physical curbs on light penetration. In contrast to optical methods, magnetic resonance imaging (MRI) may be used to noninvasively look inside intact organisms at any arbitrary depth and over large fields of view. These capabilities have spurred the development of innovative methods to connect MRI readouts with biological targets using protein-based probes that are in principle genetically encodable. Here, we highlight the state-of-the-art in MRI-based biomolecular sensors, focusing on their physical mechanisms, quantitative characteristics, and biological applications. We also describe how innovations in reporter gene technology are creating new opportunities to engineer MRI sensors that are sensitive to dilute biological targets.
Collapse
Affiliation(s)
- Jason Yun
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Michelle Baldini
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Rochishnu Chowdhury
- Mechanical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Arnab Mukherjee
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
- Center for BioEngineering, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
26
|
Nyström NN, Liu H, Martinez FM, Zhang XA, Scholl TJ, Ronald JA. Gadolinium-free Magnetic Resonance Imaging of the Liver via an Oatp1-Targeted Manganese(III) Porphyrin. J Med Chem 2022; 65:9846-9857. [PMID: 35852350 DOI: 10.1021/acs.jmedchem.2c00500] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Controversy surrounding gadolinium-based contrast agents (GBCAs) has rendered their continued utility highly contentious, but the liver-specific GBCA Gd(III) ethoxybenzyl-diethylene triamine pentaacetic acid (Gd(III)-EOB-DTPA) remains in use because it provides unique diagnostic information that could not be obtained by any other means. To address the need for an alternate liver-specific MRI probe, we synthesized Mn(III) 20-(4-ethoxyphenyl) porphyrin-5,10,15-tricarboxylate (Mn(III)TriCP-PhOEt), which exhibited significantly higher r1 relaxivity than Gd(III)-EOB-DTPA in vitro, while also targeting hepatocyte-specific organic anion-transporting polypeptide 1 (Oatp1) channels as a marker of viability. In mice, Mn(III)TriCP-PhOEt resulted in significant and specific increases in liver signal intensity on T1-weighted images and significant decreases in liver T1 time relative to pre-contrast measurements. Our findings suggest that Mn(III)TriCP-PhOEt operates as a specific and sensitive MR probe for Oatp1-targeted imaging in vivo.
Collapse
Affiliation(s)
- Nivin N Nyström
- Department of Medical Biophysics, University of Western Ontario, London N6A 3K7, Ontario, Canada
- Imaging Research Laboratories, Robarts Research Institute, Western University, London N6A 3K7, Ontario, Canada
| | - Hanlin Liu
- Department of Chemistry, University of Toronto, Toronto M5S 1A4, Ontario, Canada
- Department of Physical and Environmental Sciences, University of Toronto, Toronto M5S 1A4, Ontario, Canada
| | - Francisco M Martinez
- Imaging Research Laboratories, Robarts Research Institute, Western University, London N6A 3K7, Ontario, Canada
| | - Xiao-An Zhang
- Department of Chemistry, University of Toronto, Toronto M5S 1A4, Ontario, Canada
- Department of Physical and Environmental Sciences, University of Toronto, Toronto M5S 1A4, Ontario, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, University of Western Ontario, London N6A 3K7, Ontario, Canada
- Imaging Research Laboratories, Robarts Research Institute, Western University, London N6A 3K7, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto M5G 0A3, Ontario, Canada
| | - John A Ronald
- Department of Medical Biophysics, University of Western Ontario, London N6A 3K7, Ontario, Canada
- Imaging Research Laboratories, Robarts Research Institute, Western University, London N6A 3K7, Ontario, Canada
- Lawson Health Research Institute, London N6C 2R5, Ontario, Canada
| |
Collapse
|
27
|
Zheng N, Li M, Wu Y, Kaewborisuth C, Li Z, Gui Z, Wu J, Cai A, Lin K, Su KP, Xiang H, Tian X, Manyande A, Xu F, Wang J. A novel technology for in vivo detection of cell type-specific neural connection with AQP1-encoding rAAV2-retro vector and metal-free MRI. Neuroimage 2022; 258:119402. [PMID: 35732245 DOI: 10.1016/j.neuroimage.2022.119402] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/18/2022] [Accepted: 06/18/2022] [Indexed: 01/10/2023] Open
Abstract
A mammalian brain contains numerous neurons with distinct cell types for complex neural circuits. Virus-based circuit tracing tools are powerful in tracking the interaction among the different brain regions. However, detecting brain-wide neural networks in vivo remains challenging since most viral tracing systems rely on postmortem optical imaging. We developed a novel approach that enables in vivo detection of brain-wide neural connections based on metal-free magnetic resonance imaging (MRI). The recombinant adeno-associated virus (rAAV) with retrograde ability, the rAAV2-retro, encoding the human water channel aquaporin 1 (AQP1) MRI reporter gene was generated to label neural connections. The mouse was micro-injected with the virus at the Caudate Putamen (CPU) region and subjected to detection with Diffusion-weighted MRI (DWI). The prominent structure of the CPU-connected network was clearly defined. In combination with a Cre-loxP system, rAAV2-retro expressing Cre-dependent AQP1 provides a CPU-connected network of specific type neurons. Here, we established a sensitive, metal-free MRI-based strategy for in vivo detection of cell type-specific neural connections in the whole brain, which could visualize the dynamic changes of neural networks in rodents and potentially in non-human primates.
Collapse
Affiliation(s)
- Ning Zheng
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Mei Li
- The Brain Cognition and Brain Disease Institute (BCBDI), NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yang Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand
| | - Zhen Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhu Gui
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinfeng Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Aoling Cai
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Kangguang Lin
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kuan-Pin Su
- Department of Psychiatry, China Medical University Hospital, Taichung City, Taiwan, China
| | - Hongbing Xiang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, Middlesex, TW8 9GA, UK
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; The Brain Cognition and Brain Disease Institute (BCBDI), NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
28
|
Computationally designed dual-color MRI reporters for noninvasive imaging of transgene expression. Nat Biotechnol 2022; 40:1143-1149. [PMID: 35102291 DOI: 10.1038/s41587-021-01162-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/12/2021] [Indexed: 12/13/2022]
Abstract
Imaging of gene-expression patterns in live animals is difficult to achieve with fluorescent proteins because tissues are opaque to visible light. Imaging of transgene expression with magnetic resonance imaging (MRI), which penetrates to deep tissues, has been limited by single reporter visualization capabilities. Moreover, the low-throughput capacity of MRI limits large-scale mutagenesis strategies to improve existing reporters. Here we develop an MRI system, called GeneREFORM, comprising orthogonal reporters for two-color imaging of transgene expression in deep tissues. Starting from two promiscuous deoxyribonucleoside kinases, we computationally designed highly active, orthogonal enzymes ('reporter genes') that specifically phosphorylate two MRI-detectable synthetic deoxyribonucleosides ('reporter probes'). Systemically administered reporter probes exclusively accumulate in cells expressing the designed reporter genes, and their distribution is displayed as pseudo-colored MRI maps based on dynamic proton exchange for noninvasive visualization of transgene expression. We envision that future extensions of GeneREFORM will pave the way to multiplexed deep-tissue mapping of gene expression in live animals.
Collapse
|
29
|
Concilio SC, Russell SJ, Peng KW. A brief review of reporter gene imaging in oncolytic virotherapy and gene therapy. Mol Ther Oncolytics 2021; 21:98-109. [PMID: 33981826 PMCID: PMC8065251 DOI: 10.1016/j.omto.2021.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reporter gene imaging (RGI) can accelerate development timelines for gene and viral therapies by facilitating rapid and noninvasive in vivo studies to determine the biodistribution, magnitude, and durability of viral gene expression and/or virus infection. Functional molecular imaging systems used for this purpose can be divided broadly into deep-tissue and optical modalities. Deep-tissue modalities, which can be used in animals of any size as well as in human subjects, encompass single photon emission computed tomography (SPECT), positron emission tomography (PET), and functional/molecular magnetic resonance imaging (f/mMRI). Optical modalities encompass fluorescence, bioluminescence, Cerenkov luminescence, and photoacoustic imaging and are suitable only for small animal imaging. Here we discuss the mechanisms of action and relative merits of currently available reporter gene systems, highlighting the strengths and weaknesses of deep tissue versus optical imaging systems and the hardware/reagents that are used for data capture and processing. In light of recent technological advances, falling costs of imaging instruments, better availability of novel radioactive and optical tracers, and a growing realization that RGI can give invaluable insights across the entire in vivo translational spectrum, the approach is becoming increasingly essential to facilitate the competitive development of new virus- and gene-based drugs.
Collapse
Affiliation(s)
| | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
30
|
Farhadi A, Sigmund F, Westmeyer GG, Shapiro MG. Genetically encodable materials for non-invasive biological imaging. NATURE MATERIALS 2021; 20:585-592. [PMID: 33526879 PMCID: PMC8606175 DOI: 10.1038/s41563-020-00883-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 11/18/2020] [Indexed: 05/04/2023]
Abstract
Many questions in basic biology and medicine require the ability to visualize the function of specific cells and molecules inside living organisms. In this context, technologies such as ultrasound, optoacoustics and magnetic resonance provide non-invasive imaging access to deep-tissue regions, as used in many laboratories and clinics to visualize anatomy and physiology. In addition, recent work has enabled these technologies to image the location and function of specific cells and molecules inside the body by coupling the physics of sound waves, nuclear spins and light absorption to unique protein-based materials. These materials, which include air-filled gas vesicles, capsid-like nanocompartments, pigment-producing enzymes and transmembrane transporters, enable new forms of biomolecular and cellular contrast. The ability of these protein-based contrast agents to be genetically encoded and produced by cells creates opportunities for unprecedented in vivo studies of cellular function, while their amenability to genetic engineering enables atomic-level design of their physical, chemical and biological properties.
Collapse
Affiliation(s)
- Arash Farhadi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Felix Sigmund
- Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
- Institute for Synthetic Biomedicine, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Gil Gregor Westmeyer
- Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany.
- Institute for Synthetic Biomedicine, Helmholtz Zentrum Muenchen, Neuherberg, Germany.
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
31
|
Szucs Z, Joseph J, Larkin TJ, Xie B, Bohndiek SE, Brindle KM, Neves AA. Multi-modal imaging of high-risk ductal carcinoma in situ of the breast using C2Am: a targeted cell death imaging agent. Breast Cancer Res 2021; 23:25. [PMID: 33596961 PMCID: PMC7891030 DOI: 10.1186/s13058-021-01404-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a non-invasive form of early breast cancer, with a poorly understood natural history of invasive transformation. Necrosis is a well-recognized adverse prognostic feature of DCIS, and non-invasive detection of its presence and spatial extent could provide information not obtainable by biopsy. We describe here imaging of the distribution and extent of comedo-type necrosis in a model of human DCIS using C2Am, an imaging agent that binds to the phosphatidylserine exposed by necrotic cells. METHODS We used an established xenograft model of human DCIS that mimics the histopathological features of the disease. Planar near-infrared and optoacoustic imaging, using fluorescently labeled C2Am, were used to image non-invasively the presence and extent of lesion necrosis. RESULTS C2Am showed specific and sensitive binding to necrotic areas in DCIS tissue, detectable both in vivo and ex vivo. The imaging signal generated in vivo using near-infrared (NIR) fluorescence imaging was up to 6-fold higher in DCIS lesions than in surrounding fat pad or skin tissue. There was a correlation between the C2Am NIR fluorescence (Pearson R = 0.783, P = 0.0125) and optoacoustic signals (R > 0.875, P < 0.022) in the DCIS lesions in vivo and the corresponding levels of cell death detected histologically. CONCLUSIONS C2Am is a targeted multi-modal imaging agent that could complement current anatomical imaging methods for detecting DCIS. Imaging the presence and spatial extent of necrosis may give better prognostic information than that obtained by biopsy alone.
Collapse
Affiliation(s)
- Zoltan Szucs
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - James Joseph
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Physics, University of Cambridge, Cambridge, UK
- Present address: University of Dundee, School of Science and Engineering, Dundee, UK
| | - Tim J Larkin
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Bangwen Xie
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah E Bohndiek
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Physics, University of Cambridge, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| | - André A Neves
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
32
|
Kelly JJ, Saee-Marand M, Nyström NN, Evans MM, Chen Y, Martinez FM, Hamilton AM, Ronald JA. Safe harbor-targeted CRISPR-Cas9 homology-independent targeted integration for multimodality reporter gene-based cell tracking. SCIENCE ADVANCES 2021; 7:eabc3791. [PMID: 33523917 PMCID: PMC7817109 DOI: 10.1126/sciadv.abc3791] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/25/2020] [Indexed: 05/12/2023]
Abstract
Imaging reporter genes provides longitudinal information on the biodistribution, growth, and survival of engineered cells in vivo. A translational bottleneck to using reporter genes is the necessity to engineer cells with randomly integrating vectors. Here, we built homology-independent targeted integration (HITI) CRISPR-Cas9 minicircle donors for precise safe harbor-targeted knock-in of fluorescence, bioluminescence, and MRI (Oatp1a1) reporter genes. Our results showed greater knock-in efficiency using HITI vectors compared to homology-directed repair vectors. HITI clones demonstrated functional fluorescence and bioluminescence reporter activity as well as significant Oatp1a1-mediated uptake of the clinically approved MRI agent gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid. Contrast-enhanced MRI improved the conspicuity of both subcutaneous and metastatic Oatp1a1-expressing tumors before they became palpable or even readily visible on precontrast images. Our work demonstrates the first CRISPR-Cas9 HITI system for knock-in of large DNA donor constructs at a safe harbor locus, enabling multimodal longitudinal in vivo imaging of cells.
Collapse
Affiliation(s)
- John J Kelly
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Moe Saee-Marand
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Nivin N Nyström
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Melissa M Evans
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Yuanxin Chen
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Francisco M Martinez
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Amanda M Hamilton
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - John A Ronald
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
33
|
Jacobs AH, Schelhaas S, Viel T, Waerzeggers Y, Winkeler A, Zinnhardt B, Gelovani J. Imaging of Gene and Cell-Based Therapies: Basis and Clinical Trials. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
34
|
Bulat F, Hesse F, Hu DE, Ros S, Willminton-Holmes C, Xie B, Attili B, Soloviev D, Aigbirhio F, Leeper FJ, Brindle KM, Neves AA. 18F-C2Am: a targeted imaging agent for detecting tumor cell death in vivo using positron emission tomography. EJNMMI Res 2020; 10:151. [PMID: 33296043 PMCID: PMC7726082 DOI: 10.1186/s13550-020-00738-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Trialing novel cancer therapies in the clinic would benefit from imaging agents that can detect early evidence of treatment response. The timing, extent and distribution of cell death in tumors following treatment can give an indication of outcome. We describe here an 18F-labeled derivative of a phosphatidylserine-binding protein, the C2A domain of Synaptotagmin-I (C2Am), for imaging tumor cell death in vivo using PET. METHODS A one-pot, two-step automated synthesis of N-(5-[18F]fluoropentyl)maleimide (60 min synthesis time, > 98% radiochemical purity) has been developed, which was used to label the single cysteine residue in C2Am within 30 min at room temperature. Binding of 18F-C2Am to apoptotic and necrotic tumor cells was assessed in vitro, and also in vivo, by dynamic PET and biodistribution measurements in mice bearing human tumor xenografts treated with a TRAILR2 agonist or with conventional chemotherapy. C2Am detection of tumor cell death was validated by correlation of probe binding with histological markers of cell death in tumor sections obtained immediately after imaging. RESULTS 18F-C2Am showed a favorable biodistribution profile, with predominantly renal clearance and minimal retention in spleen, liver, small intestine, bone and kidney, at 2 h following probe administration. 18F-C2Am generated tumor-to-muscle (T/m) ratios of 6.1 ± 2.1 and 10.7 ± 2.4 within 2 h of probe administration in colorectal and breast tumor models, respectively, following treatment with the TRAILR2 agonist. The levels of cell death (CC3 positivity) following treatment were 12.9-58.8% and 11.3-79.7% in the breast and colorectal xenografts, respectively. Overall, a 20% increase in CC3 positivity generated a one unit increase in the post/pre-treatment tumor contrast. Significant correlations were found between tracer uptake post-treatment, at 2 h post-probe administration, and histological markers of cell death (CC3: Pearson R = 0.733, P = 0.0005; TUNEL: Pearson R = 0.532, P = 0.023). CONCLUSION The rapid clearance of 18F-C2Am from the blood pool and low kidney retention allowed the spatial distribution of cell death in a tumor to be imaged during the course of therapy, providing a rapid assessment of tumor treatment response. 18F-C2Am has the potential to be used in the clinic to assess early treatment response in tumors.
Collapse
Affiliation(s)
- Flaviu Bulat
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Friederike Hesse
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - De-En Hu
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Susana Ros
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | | | - Bangwen Xie
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Bala Attili
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Dmitry Soloviev
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Franklin Aigbirhio
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Finian J Leeper
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - André A Neves
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
35
|
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: 2.2] [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
|
36
|
Baek SE, Ul-Haq A, Kim DH, Choi HW, Kim MJ, Choi HJ, Kim H. Human Organic Anion Transporting Polypeptide 1B3 Applied as an MRI-Based Reporter Gene. Korean J Radiol 2020; 21:726-735. [PMID: 32410411 PMCID: PMC7231618 DOI: 10.3348/kjr.2019.0903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/19/2020] [Indexed: 12/22/2022] Open
Abstract
Objective Recent innovations in biology are boosting gene and cell therapy, but monitoring the response to these treatments is difficult. The purpose of this study was to find an MRI-reporter gene that can be used to monitor gene or cell therapy and that can be delivered without a viral vector, as viral vector delivery methods can result in long-term complications. Materials and Methods CMV promoter-human organic anion transporting polypeptide 1B3 (CMV-hOATP1B3) cDNA or CMV-blank DNA (control) was transfected into HEK293 cells using Lipofectamine. OATP1B3 expression was confirmed by western blotting and confocal microscopy. In vitro cell phantoms were made using transfected HEK293 cells cultured in various concentrations of gadoxetic acid for 24 hours, and images of the phantoms were made with a 9.4T micro-MRI. In vivo xenograft tumors were made by implanting HEK293 cells transfected with CMV-hOATP1B3 (n = 4) or CMV-blank (n = 4) in 8-week-old male nude mice, and MRI was performed before and after intravenous injection of gadoxetic acid (1.2 µL/g). Results Western blot and confocal microscopy after immunofluorescence staining revealed that only CMV-hOATP1B3-transfected HEK293 cells produced abundant OATP1B3, which localized at the cell membrane. OATP1B3 expression levels remained high through the 25th subculture cycle, but decreased substantially by the 50th subculture cycle. MRI of cell phantoms showed that only the CMV-hOATP1B3-transfected cells produced a significant contrast enhancement effect. In vivo MRI of xenograft tumors revealed that only CMV-hOATP1B3-transfected HEK293 tumors demonstrated a T1 contrast effect, which lasted for at least 5 hours. Conclusion The human endogenous OATP1B3 gene can be non-virally delivered into cells to induce transient OATP1B3 expression, leading to gadoxetic acid-mediated enhancement on MRI. These results indicate that hOATP1B3 can serve as an MRI-reporter gene while minimizing the risk of long-term complications.
Collapse
Affiliation(s)
- Song Ee Baek
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Asad Ul-Haq
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Hee Kim
- Yonsei University College of Medicine, Seoul, Korea
| | | | - Myeong Jin Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jin Choi
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Honsoul Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
37
|
Patrick PS, Kolluri KK, Zaw Thin M, Edwards A, Sage EK, Sanderson T, Weil BD, Dickson JC, Lythgoe MF, Lowdell M, Janes SM, Kalber TL. Lung delivery of MSCs expressing anti-cancer protein TRAIL visualised with 89Zr-oxine PET-CT. Stem Cell Res Ther 2020; 11:256. [PMID: 32586403 PMCID: PMC7318529 DOI: 10.1186/s13287-020-01770-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/01/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND MSCTRAIL is a cell-based therapy consisting of human allogeneic umbilical cord-derived MSCs genetically modified to express the anti-cancer protein TRAIL. Though cell-based therapies are typically designed with a target tissue in mind, delivery is rarely assessed due to a lack of translatable non-invasive imaging approaches. In this preclinical study, we demonstrate 89Zr-oxine labelling and PET-CT imaging as a potential clinical solution for non-invasively tracking MSCTRAIL biodistribution. Future implementation of this technique should improve our understanding of MSCTRAIL during its evaluation as a therapy for metastatic lung adenocarcinoma. METHODS MSCTRAIL were radiolabelled with 89Zr-oxine and assayed for viability, phenotype, and therapeutic efficacy post-labelling. PET-CT imaging of 89Zr-oxine-labelled MSCTRAIL was performed in a mouse model of lung cancer following intravenous injection, and biodistribution was confirmed ex vivo. RESULTS MSCTRAIL retained the therapeutic efficacy and MSC phenotype in vitro at labelling amounts up to and above those required for clinical imaging. The effect of 89Zr-oxine labelling on cell proliferation rate was amount- and time-dependent. PET-CT imaging showed delivery of MSCTRAIL to the lungs in a mouse model of lung cancer up to 1 week post-injection, validated by in vivo bioluminescence imaging, autoradiography, and fluorescence imaging on tissue sections. CONCLUSIONS 89Zr-oxine labelling and PET-CT imaging present a potential method of evaluating the biodistribution of new cell therapies in patients, including MSCTRAIL. This offers to improve understanding of cell therapies, including mechanism of action, migration dynamics, and inter-patient variability.
Collapse
Affiliation(s)
- P Stephen Patrick
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK.
| | - Krishna K Kolluri
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - May Zaw Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - Adam Edwards
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Elizabeth K Sage
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Tom Sanderson
- Institute of Nuclear Medicine, University College London, London, UK
| | - Benjamin D Weil
- Centre for Cell, Gene & Tissue Therapeutics, Royal Free Hospital, London, UK
| | - John C Dickson
- Institute of Nuclear Medicine, University College London, London, UK
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - Mark Lowdell
- Centre for Cell, Gene & Tissue Therapeutics, Royal Free Hospital, London, UK
- Department of Haematology, Cancer Institute, University College London, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK.
| |
Collapse
|
38
|
Abstract
Topical drug delivery has inherent advantages over other administration routes. However, the existence of stratum corneum limits the diffusion to small and lipophilic drugs. Fortunately, the advancement of nanotechnology brings along opportunities to address this challenge. Taking the unique features in size and surface chemistry, nanocarriers such as liposomes, polymeric nanoparticles, gold nanoparticles, and framework nucleic acids have been used to bring drugs across the skin barrier to epidermis and dermis layers. This article reviews the development of these formulations and focuses on their applications in the treatment of skin disorders such as acne, skin inflammation, skin infection, and wound healing. Existing hurdles and further developments are also discussed.
Collapse
Affiliation(s)
- Mingyue Cui
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457
| | - Sharon Wan Ting Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457.,National Dental Centre of Singapore, 5 Second Hospital Avenue, Singapore 168938.,Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| |
Collapse
|
39
|
Serganova I, Blasberg RG. Molecular Imaging with Reporter Genes: Has Its Promise Been Delivered? J Nucl Med 2020; 60:1665-1681. [PMID: 31792128 DOI: 10.2967/jnumed.118.220004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
The first reporter systems were developed in the early 1980s and were based on measuring the activity of an enzyme-as a surrogate measure of promoter-driven transcriptional activity-which is now known as a reporter gene system. The initial objective and application of reporter techniques was to analyze the activity of a specific promoter (namely, the expression of a gene that is under the regulation of the specific promoter that is linked to the reporter gene). This system allows visualization of specific promoter activity with great sensitivity. In general, there are 2 classes of reporter systems: constitutively expressed (always-on) reporter constructs used for cell tracking, and inducible reporter systems sensitive to endogenous signaling molecules and transcription factors that characterize specific tissues, tumors, or signaling pathways.This review traces the development of different reporter systems, using fluorescent and bioluminescent proteins as well as radionuclide-based reporter systems. The development and application of radionuclide-based reporter systems is the focus of this review. The question at the end of the review is whether the "promise" of reporter gene imaging has been realized. What is required for moving forward with radionuclide-based reporter systems, and what is required for successful translation to clinical applications?
Collapse
Affiliation(s)
- Inna Serganova
- Department of Neurology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald G Blasberg
- Department of Neurology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Memorial Hospital, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
40
|
Pan CT, Chang WH, Kumar A, Singh SP, Kaushik AC, Sharma J, Long ZJ, Wen ZH, Mishra SK, Yen CK, Chaudhary RK, Shiue YL. Nanoparticles-mediated Brain Imaging and Disease Prognosis by Conventional as well as Modern Modal Imaging Techniques: a Comparison. Curr Pharm Des 2020; 25:2637-2649. [PMID: 31603057 DOI: 10.2174/1381612825666190709220139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Multimodal imaging plays an important role in the diagnosis of brain disorders. Neurological disorders need to be diagnosed at an early stage for their effective treatment as later, it is very difficult to treat them. If possible, diagnosing at an early stage can be much helpful in curing the disease with less harm to the body. There is a need for advanced and multimodal imaging techniques for the same. This paper provides an overview of conventional as well as modern imaging techniques for brain diseases, specifically for tumor imaging. In this paper, different imaging modalities are discussed for tumor detection in the brain along with their advantages and disadvantages. Conjugation of two and more than two modalities provides more accurate information rather than a single modality. They can monitor and differentiate the cellular processes of normal and diseased condition with more clarity. The advent of molecular imaging, including reporter gene imaging, has opened the door of more advanced noninvasive detection of brain tumors. Due to specific optical properties, semiconducting polymer-based nanoparticles also play a pivotal role in imaging tumors. OBJECTIVE The objective of this paper is to review nanoparticles-mediated brain imaging and disease prognosis by conventional as well as modern modal imaging techniques. CONCLUSION We reviewed in detail various medical imaging techniques. This paper covers recent developments in detail and elaborates a possible research aspect for the readers in the field.
Collapse
Affiliation(s)
- Cheng-Tang Pan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Wei-Hsi Chang
- Department of Emergency Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Ajay Kumar
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Satya P Singh
- School of EEE, Nanyang Technological University, Nanyang Ave, Singapore
| | - Aman Chandra Kaushik
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, ShanghaiJia Tong University, Shanghai 200240, China
| | - Jyotsna Sharma
- Amity School of Applied Sciences, Amity University Haryana, Gurugram-122413, Manesai, Panchgaon, Haryana, India
| | - Zheng-Jing Long
- Department of Emergency Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Sunil Kumar Mishra
- Patronage Institute of Management Studies, Greater Noida, Uttar Pradesh, India
| | - Chung-Kun Yen
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Ravi Kumar Chaudhary
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pardesh, India, India
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| |
Collapse
|
41
|
Hu H. Recent Advances of Bioresponsive Nano-Sized Contrast Agents for Ultra-High-Field Magnetic Resonance Imaging. Front Chem 2020; 8:203. [PMID: 32266217 PMCID: PMC7100386 DOI: 10.3389/fchem.2020.00203] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022] Open
Abstract
The ultra-high-field magnetic resonance imaging (MRI) nowadays has been receiving enormous attention in both biomaterial research and clinical diagnosis. MRI contrast agents are generally comprising of T1-weighted and T2-weighted contrast agent types, where T1-weighted contrast agents show positive contrast enhancement with brighter images by decreasing the proton's longitudinal relaxation times and T2-weighted contrast agents show negative contrast enhancement with darker images by decreasing the proton's transverse relaxation times. To meet the incredible demand of MRI, ultra-high-field T2 MRI is gradually attracting the attention of research and medical needs owing to its high resolution and high accuracy for detection. It is anticipated that high field MRI contrast agents can achieve high performance in MRI imaging, where parameters of chemical composition, molecular structure and size of varied contrast agents show contrasted influence in each specific diagnostic test. This review firstly presents the recent advances of nanoparticle contrast agents for MRI. Moreover, multimodal molecular imaging with MRI for better monitoring is discussed during biological process. To fasten the process of developing better contrast agents, deep learning of artificial intelligent (AI) can be well-integrated into optimizing the crucial parameters of nanoparticle contrast agents and achieving high resolution MRI prior to the clinical applications. Finally, prospects and challenges are summarized.
Collapse
Affiliation(s)
- Hailong Hu
- School of Aeronautics and Astronautics, Central South University, Changsha, China
- Research Center in Intelligent Thermal Structures for Aerospace, Central South University, Changsha, China
| |
Collapse
|
42
|
Ozbakir HF, Anderson NT, Fan KC, Mukherjee A. Beyond the Green Fluorescent Protein: Biomolecular Reporters for Anaerobic and Deep-Tissue Imaging. Bioconjug Chem 2020; 31:293-302. [PMID: 31794658 PMCID: PMC7033020 DOI: 10.1021/acs.bioconjchem.9b00688] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fluorescence imaging represents cornerstone technology for studying biological function at the cellular and molecular levels. The technology's centerpiece is a prolific collection of genetic reporters based on the green fluorescent protein (GFP) and related analogs. More than two decades of protein engineering have endowed the GFP repertoire with an incredible assortment of fluorescent proteins, allowing scientists immense latitude in choosing reporters tailored to various cellular and environmental contexts. Nevertheless, GFP and derivative reporters have specific limitations that hinder their unrestricted use for molecular imaging. These challenges have inspired the development of new reporter proteins and imaging mechanisms. Here, we review how these developments are expanding the frontiers of reporter gene techniques to enable nondestructive studies of cell function in anaerobic environments and deep inside intact animals-two important biological contexts that are fundamentally incompatible with the use of GFP-based reporters.
Collapse
Affiliation(s)
- Harun F. Ozbakir
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Nolan T. Anderson
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Kang-Ching Fan
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Arnab Mukherjee
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry, University of California, Santa Barbara, California 93106, United States
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
- Center for Bioengineering, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
43
|
Hernández Lozano I, Langer O. Use of imaging to assess the activity of hepatic transporters. Expert Opin Drug Metab Toxicol 2020; 16:149-164. [PMID: 31951754 PMCID: PMC7055509 DOI: 10.1080/17425255.2020.1718107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Introduction: Membrane transporters of the SLC and ABC families are abundantly expressed in the liver, where they control the transfer of drugs/drug metabolites across the sinusoidal and canalicular hepatocyte membranes and play a pivotal role in hepatic drug clearance. Noninvasive imaging methods, such as PET, SPECT or MRI, allow for measuring the activity of hepatic transporters in vivo, provided that suitable transporter imaging probes are available.Areas covered: We give an overview of the working principles of imaging-based assessment of hepatic transporter activity. We discuss different currently available PET/SPECT radiotracers and MRI contrast agents and their applications to measure hepatic transporter activity in health and disease. We cover mathematical modeling approaches to obtain quantitative parameters of transporter activity and provide a critical assessment of methodological limitations and challenges associated with this approach.Expert opinion: PET in combination with pharmacokinetic modeling can be potentially applied in drug development to study the distribution of new drug candidates to the liver and their clearance mechanisms. This approach bears potential to mechanistically assess transporter-mediated drug-drug interactions, to assess the influence of disease on hepatic drug disposition and to validate and refine currently available in vitro-in vivo extrapolation methods to predict hepatic clearance of drugs.
Collapse
Affiliation(s)
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
44
|
Longitudinal Visualization of Viable Cancer Cell Intratumoral Distribution in Mouse Models Using Oatp1a1-Enhanced Magnetic Resonance Imaging. Invest Radiol 2019; 54:302-311. [PMID: 30672844 DOI: 10.1097/rli.0000000000000542] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Multimodality reporter gene imaging provides valuable, noninvasive information on the fate of engineered cell populations. To complement magnetic resonance imaging (MRI) measures of tumor volume and 2-dimensional reporter-based optical measures of cell viability, reporter-based MRI may offer 3-dimensional information on the distribution of viable cancer cells in deep tissues. MATERIALS AND METHODS Here, we engineered human and murine triple-negative breast cancer cells with lentivirus encoding tdTomato and firefly luciferase for fluorescence imaging and bioluminescence imaging (BLI). A subset of these cells was additionally engineered with lentivirus encoding organic anion transporting polypeptide 1a1 (Oatp1a1) for MRI. Oatp1a1 operates by transporting gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA) into cells, and it concomitantly improves BLI substrate uptake. After orthotopic implantation of engineered cells expressing or not expressing Oatp1a1, longitudinal fluorescence imaging, BLI, and 3-Tesla MRI were performed. RESULTS Oatp1a1-expressing tumors displayed significantly increased BLI signals relative to control tumors at all time points (P < 0.05). On MRI, post-Gd-EOB-DTPA T1-weighted images of Oatp1a1-expressing tumors exhibited significantly increased contrast-to-noise ratios compared with control tumors and precontrast images (P < 0.05). At endpoint, tumors expressing Oatp1a1 displayed intratumoral MR signal heterogeneity not present at earlier time points. Pixel-based analysis of matched in vivo MR and ex vivo fluorescence microscopy images revealed a strong, positive correlation between MR intensity and tdTomato intensity for Oatp1a1-expressing tumors (P < 0.05), but not control tumors. CONCLUSIONS These results characterize Oatp1a1 as a sensitive, quantitative, positive contrast MRI reporter gene for 3-dimensional assessment of viable cancer cell intratumoral distribution and concomitant BLI enhancement. This multimodality reporter gene system can provide new insights into the influence of viable cancer cell intratumoral distribution on tumor progression and metastasis, as well as improved assessments of anticancer therapies.
Collapse
|
45
|
Nyström NN, Yip LCM, Carson JJL, Scholl TJ, Ronald JA. Development of a Human Photoacoustic Imaging Reporter Gene Using the Clinical Dye Indocyanine Green. Radiol Imaging Cancer 2019; 1:e190035. [PMID: 33778683 DOI: 10.1148/rycan.2019190035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/17/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022]
Abstract
Purpose To develop a photoacoustic imaging (PAI) reporter gene that has high translational potential. Previous research has shown that human organic anion-transporting polypeptide 1b3 (OATP1B3) promotes the uptake of the near-infrared fluorescent dye indocyanine green (ICG). In this study, the authors have established OATP1B3 and ICG as a reporter gene-probe pair for in vivo PAI. Materials and Methods Human breast cancer cells were engineered to express OATP1B3. Control cells (not expressing OATP1B3) or OATP1B3-expressing cells were incubated with or without ICG, placed in a breast-mimicking phantom, and imaged with PAI. Control (n = 6) or OATP1B3-expressing (n = 5) cells were then implanted orthotopically into female mice. Full-spectrum PAI was performed before and 24 hours after ICG administration. One-way analysis of variance was performed, followed by Tukey posthoc multiple comparisons, to assess statistical significance. Results OATP1B3-expressing cells incubated with ICG exhibited a 2.7-fold increase in contrast-to-noise ratio relative to all other controls in vitro (P < .05). In mice, PAI signals after ICG administration were increased 2.3-fold in OATP1B3 tumors relative to those in controls (P < .05). Conclusion OATP1B3 operates as an in vivo PAI reporter gene based on its ability to promote the cellular uptake of ICG. Benefits include the human derivation of OATP1B3, combined with the use of wavelengths in the near-infrared region, high extinction coefficient, low quantum yield, and clinical approval of ICG. The authors posit that this system will be useful for localized monitoring of emerging gene- and cell-based therapies in clinical applications.© RSNA, 2019Keywords: Animal Studies, Molecular Imaging, Molecular Imaging-Clinical Translation, Molecular Imaging-Reporter Gene Imaging, Optical ImagingSupplemental material is available for this article.
Collapse
Affiliation(s)
- Nivin N Nyström
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond St N, Room 2241A, London, ON, Canada N6A 3K7 (N.N.N., L.C.M.Y., J.J.L.C., T.J.S., J.A.R.); Imaging Research Laboratories, Robarts Research Institute, London, Canada (N.N.N., T.J.S., J.A.R.); Lawson Health Research Institute, London, Canada (L.C.M.Y., J.J.L.C., J.A.R.); and Ontario Institute for Cancer Research, Toronto, Canada (T.J.S.)
| | - Lawrence C M Yip
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond St N, Room 2241A, London, ON, Canada N6A 3K7 (N.N.N., L.C.M.Y., J.J.L.C., T.J.S., J.A.R.); Imaging Research Laboratories, Robarts Research Institute, London, Canada (N.N.N., T.J.S., J.A.R.); Lawson Health Research Institute, London, Canada (L.C.M.Y., J.J.L.C., J.A.R.); and Ontario Institute for Cancer Research, Toronto, Canada (T.J.S.)
| | - Jeffrey J L Carson
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond St N, Room 2241A, London, ON, Canada N6A 3K7 (N.N.N., L.C.M.Y., J.J.L.C., T.J.S., J.A.R.); Imaging Research Laboratories, Robarts Research Institute, London, Canada (N.N.N., T.J.S., J.A.R.); Lawson Health Research Institute, London, Canada (L.C.M.Y., J.J.L.C., J.A.R.); and Ontario Institute for Cancer Research, Toronto, Canada (T.J.S.)
| | - Timothy J Scholl
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond St N, Room 2241A, London, ON, Canada N6A 3K7 (N.N.N., L.C.M.Y., J.J.L.C., T.J.S., J.A.R.); Imaging Research Laboratories, Robarts Research Institute, London, Canada (N.N.N., T.J.S., J.A.R.); Lawson Health Research Institute, London, Canada (L.C.M.Y., J.J.L.C., J.A.R.); and Ontario Institute for Cancer Research, Toronto, Canada (T.J.S.)
| | - John A Ronald
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond St N, Room 2241A, London, ON, Canada N6A 3K7 (N.N.N., L.C.M.Y., J.J.L.C., T.J.S., J.A.R.); Imaging Research Laboratories, Robarts Research Institute, London, Canada (N.N.N., T.J.S., J.A.R.); Lawson Health Research Institute, London, Canada (L.C.M.Y., J.J.L.C., J.A.R.); and Ontario Institute for Cancer Research, Toronto, Canada (T.J.S.)
| |
Collapse
|
46
|
Hesketh RL, Wang J, Wright AJ, Lewis DY, Denton AE, Grenfell R, Miller JL, Bielik R, Gehrung M, Fala M, Ros S, Xie B, Hu DE, Brindle KM. Magnetic Resonance Imaging Is More Sensitive Than PET for Detecting Treatment-Induced Cell Death-Dependent Changes in Glycolysis. Cancer Res 2019; 79:3557-3569. [PMID: 31088837 PMCID: PMC6640042 DOI: 10.1158/0008-5472.can-19-0182] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/30/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022]
Abstract
Metabolic imaging has been widely used to measure the early responses of tumors to treatment. Here, we assess the abilities of PET measurement of [18F]FDG uptake and MRI measurement of hyperpolarized [1-13C]pyruvate metabolism to detect early changes in glycolysis following treatment-induced cell death in human colorectal (Colo205) and breast adenocarcinoma (MDA-MB-231) xenografts in mice. A TRAIL agonist that binds to human but not mouse cells induced tumor-selective cell death. Tumor glycolysis was assessed by injecting [1,6-13C2]glucose and measuring 13C-labeled metabolites in tumor extracts. Injection of hyperpolarized [1-13C]pyruvate induced rapid reduction in lactate labeling. This decrease, which correlated with an increase in histologic markers of cell death and preceded decrease in tumor volume, reflected reduced flux from glucose to lactate and decreased lactate concentration. However, [18F]FDG uptake and phosphorylation were maintained following treatment, which has been attributed previously to increased [18F]FDG uptake by infiltrating immune cells. Quantification of [18F]FDG uptake in flow-sorted tumor and immune cells from disaggregated tumors identified CD11b+/CD45+ macrophages as the most [18F]FDG-avid cell type present, yet they represented <5% of the cells present in the tumors and could not explain the failure of [18F]FDG-PET to detect treatment response. MRI measurement of hyperpolarized [1-13C]pyruvate metabolism is therefore a more sensitive marker of the early decreases in glycolytic flux that occur following cell death than PET measurements of [18F]FDG uptake. SIGNIFICANCE: These findings demonstrate superior sensitivity of MRI measurement of hyperpolarized [1-13C]pyruvate metabolism versus PET measurement of 18F-FDG uptake for detecting early changes in glycolysis following treatment-induced tumor cell death.
Collapse
Affiliation(s)
- Richard L Hesketh
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Jiazheng Wang
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Alan J Wright
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - David Y Lewis
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Alice E Denton
- Lymphocyte Signalling and Development, Babraham Hall House, Babraham, Cambridge, United Kingdom
| | - Richard Grenfell
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Jodi L Miller
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Robert Bielik
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Marcel Gehrung
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Maria Fala
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Susana Ros
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Bangwen Xie
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - De-En Hu
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Kevin M Brindle
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom.
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
47
|
Wu MR, Huang YY, Hsiao JK. Use of Indocyanine Green (ICG), a Medical Near Infrared Dye, for Enhanced Fluorescent Imaging-Comparison of Organic Anion Transporting Polypeptide 1B3 (OATP1B3) and Sodium-Taurocholate Cotransporting Polypeptide (NTCP) Reporter Genes. Molecules 2019; 24:molecules24122295. [PMID: 31234288 PMCID: PMC6630416 DOI: 10.3390/molecules24122295] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Molecular and cellular imaging in living organisms have ushered in an era of comprehensive understanding of intracellular and intercellular events. Currently, more efforts have been focused on the infrared fluorescent dyes that facilitate deeper tissue visualization. Both sodium taurocholate cotransporting polypeptide (NTCP) and organic-anion-transporting polypeptide 1B3 (OATP1B3) are capable of carrying indocyanine green (ICG) into the cytoplasm. We compared the feasibility of NTCP and OATP1B3 as reporter genes in combination with ICG. NTCP and OATP1B3 were transduced into HT-29 cells. Genetically modified HT-29 cells were inoculated into nude mice. ICG was administered in vitro and in vivo and the signals were observed under confocal microscopy, flow cytometry, multimode microplate reader, and an in vivo imaging system. Both NTCP- and OATP1B3-expressing cells and xenografts had higher ICG intensities. The OATP1B3-expressing xenograft has a higher ICG uptake than the NTCP-expressing xenograft. NTCP or OATP1B3 combined with ICG could serve as a noninvasive imaging modality for molecular and cellular imaging. OATP1B3 outperforms NTCP in terms of in vivo imaging.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan.
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei city 23142, Taiwan.
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan.
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei city 23142, Taiwan.
- School of Medicine, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd. Hualien 97004, Taiwan.
| |
Collapse
|
48
|
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: 4] [Impact Index Per Article: 0.7] [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
|
49
|
Wahsner J, Gale EM, Rodríguez-Rodríguez A, Caravan P. Chemistry of MRI Contrast Agents: Current Challenges and New Frontiers. Chem Rev 2019; 119:957-1057. [PMID: 30350585 PMCID: PMC6516866 DOI: 10.1021/acs.chemrev.8b00363] [Citation(s) in RCA: 905] [Impact Index Per Article: 150.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tens of millions of contrast-enhanced magnetic resonance imaging (MRI) exams are performed annually around the world. The contrast agents, which improve diagnostic accuracy, are almost exclusively small, hydrophilic gadolinium(III) based chelates. In recent years concerns have arisen surrounding the long-term safety of these compounds, and this has spurred research into alternatives. There has also been a push to develop new molecularly targeted contrast agents or agents that can sense pathological changes in the local environment. This comprehensive review describes the state of the art of clinically approved contrast agents, their mechanism of action, and factors influencing their safety. From there we describe different mechanisms of generating MR image contrast such as relaxation, chemical exchange saturation transfer, and direct detection and the types of molecules that are effective for these purposes. Next we describe efforts to make safer contrast agents either by increasing relaxivity, increasing resistance to metal ion release, or by moving to gadolinium(III)-free alternatives. Finally we survey approaches to make contrast agents more specific for pathology either by direct biochemical targeting or by the design of responsive or activatable contrast agents.
Collapse
Affiliation(s)
- Jessica Wahsner
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Aurora Rodríguez-Rodríguez
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
50
|
Takayama Y, Kusamori K, Nishikawa M. Click Chemistry as a Tool for Cell Engineering and Drug Delivery. Molecules 2019; 24:molecules24010172. [PMID: 30621193 PMCID: PMC6337375 DOI: 10.3390/molecules24010172] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/14/2023] Open
Abstract
Click chemistry has great potential for use in binding between nucleic acids, lipids, proteins, and other molecules, and has been used in many research fields because of its beneficial characteristics, including high yield, high specificity, and simplicity. The recent development of copper-free and less cytotoxic click chemistry reactions has allowed for the application of click chemistry to the field of medicine. Moreover, metabolic glycoengineering allows for the direct modification of living cells with substrates for click chemistry either in vitro or in vivo. As such, click chemistry has become a powerful tool for cell transplantation and drug delivery. In this review, we describe some applications of click chemistry for cell engineering in cell transplantation and for drug delivery in the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|