1
|
Bejoy J, Welch RC, Qian ES, Williams FM, Gibson-Corley KN, Wilson MH, Paragas N, Woodard LE. Urine-derived stem cells display homing, incorporation, and regeneration in human organoid and mouse models of acute kidney injury. Mol Ther 2025:S1525-0016(25)00219-9. [PMID: 40158205 DOI: 10.1016/j.ymthe.2025.03.044] [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: 06/27/2024] [Revised: 11/03/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Urine-derived stem cells (USCs) are adult human stem cells that can be collected noninvasively from urine and cultured in vitro. Because of their renal origin and reported therapeutic effects, we hypothesized that USCs would home to the injured kidney in acute kidney injury (AKI) models. We used mouse models of glycerol-induced rhabdomyolysis or unilateral nephrectomy with clamping ischemia reperfusion injury to model AKI. To track USC homing by live animal imaging, we administered luciferase-expressing (Luc) USCs to mice by intraperitoneal injection. We observed USC localization to both the tubules and glomeruli of injured mice within 3 h by histology. We confirmed the presence of Luc-USCs in the kidney at 3 h, 24 h, and 48 h after the injection using biodistribution analysis of quantitative bioluminescence tomography imaging. We performed immunostaining for kidney injury molecule-1 (KIM-1/HAVCR1/TIM-1) for kidney injury and found reduced expression in USC-treated group at 24 h after injection. To evaluate the effects of the human USCs on injured human nephrons, we injured human kidney organoids with the nephrotoxin cisplatin (5 μM) followed by 5 × 104 USC treatment. USCs were incorporated and lowered expression of KIM-1 in the organoids. USCs home to injured nephrons and reduce measures of kidney injury.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Richard C Welch
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Eddie S Qian
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Felisha M Williams
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katherine N Gibson-Corley
- Department of Medicine, Division of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Comparative Pathology and Research Histology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew H Wilson
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Nashville, TN 37212, USA; Departments of Pharmacology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Neal Paragas
- Department of Radiology, University of Washington, Seattle, WA 98109, USA
| | - Lauren E Woodard
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Nashville, TN 37212, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
2
|
Li J, Wang Z, Wei Y, Li W, He M, Kang J, Xu J, Liu D. Advances in Tracing Techniques: Mapping the Trajectory of Mesenchymal Stem-Cell-Derived Extracellular Vesicles. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:137-168. [PMID: 40151822 PMCID: PMC11938168 DOI: 10.1021/cbmi.4c00085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 03/29/2025]
Abstract
Mesenchymal stem-cell-derived extracellular vesicles (MSC-EVs) are nanoscale lipid bilayer vesicles secreted by mesenchymal stem cells. They inherit the parent cell's attributes, facilitating tissue repair and regeneration, promoting angiogenesis, and modulating the immune response, while offering advantages like reduced immunogenicity, straightforward administration, and enhanced stability for long-term storage. These characteristics elevate MSC-EVs as highly promising in cell-free therapy with notable clinical potential. It is critical to delve into their pharmacokinetics and thoroughly elucidate their intracellular and in vivo trajectories. A detailed summary and evaluation of existing tracing strategies are needed to establish standardized protocols. Here, we have summarized and anticipated the research progress of MSC-EVs in various biomedical imaging techniques, including fluorescence imaging, bioluminescence imaging, nuclear imaging (PET, SPECT), tomographic imaging (CT, MRI), and photoacoustic imaging. The challenges and prospects of MSC-EV tracing strategies, with particular emphasis on clinical translation, have been analyzed, with promising solutions proposed.
Collapse
Affiliation(s)
- Jingqi Li
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhaoyu Wang
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yongchun Wei
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Wenshuai Li
- State
Key Laboratory for Crop Stress Resistance and High-Efficiency Production,
Shaanxi Key Laboratory of Agricultural and Environmental Microbiology,
College of Life Sciences, Northwest A&F
University, Yangling, Shaanxi 712100, China
| | - Mingzhu He
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jingjing Kang
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jia Xu
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dingbin Liu
- State
Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory
of Molecular Recognition and Biosensing, Frontiers Science Centers
for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
3
|
Deng Z, Xu X, Dehghani H, Reyes J, Zheng L, Tran PT, Wang KKH. In vivo bioluminescence tomography-guided system for pancreatic cancer radiotherapy research. BIOMEDICAL OPTICS EXPRESS 2024; 15:4525-4539. [PMID: 39347008 PMCID: PMC11427198 DOI: 10.1364/boe.523916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 10/01/2024]
Abstract
Recent development of radiotherapy (RT) has heightened the use of radiation in managing pancreatic cancer. Thus, there is a need to investigate pancreatic cancer in a pre-clinical setting to advance our understanding of the role of RT. Widely-used cone-beam CT (CBCT) imaging cannot provide sufficient soft tissue contrast to guide irradiation. The pancreas is also prone to motion. Large collimation is unavoidably used for irradiation, costing normal tissue toxicity. We innovated a bioluminescence tomography (BLT)-guided system to address these needs. We established an orthotopic pancreatic ductal adenocarcinoma (PDAC) mouse model to access BLT. Mice underwent multi-projection and multi-spectral bioluminescence imaging (BLI), followed by CBCT imaging in an animal irradiator for BLT reconstruction and radiation planning. With optimized absorption coefficients, BLT localized PDAC at 1.25 ± 0.19 mm accuracy. To account for BLT localization uncertainties, we expanded the BLT-reconstructed volume with margin to form planning target volume(PTVBLT) for radiation planning, covering 98.7 ± 2.2% of PDAC. The BLT-guided conformal plan can cover 100% of tumors with limited normal tissue involvement across both inter-animal and inter-fraction cases, superior to the 2D BLI-guided conventional plan. BLT offers unique opportunities to localize PDAC for conformal irradiation, minimize normal tissue involvement, and support reproducibility in RT studies.
Collapse
Affiliation(s)
- Zijian Deng
- Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Xiangkun Xu
- Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| | - Hamid Dehghani
- School of Computer Science, University of Birmingham, Birmingham B15 2TT, USA
| | - Juvenal Reyes
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Ken Kang-Hsin Wang
- Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
4
|
Androsavich JR. Frameworks for transformational breakthroughs in RNA-based medicines. Nat Rev Drug Discov 2024; 23:421-444. [PMID: 38740953 DOI: 10.1038/s41573-024-00943-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/16/2024]
Abstract
RNA has sparked a revolution in modern medicine, with the potential to transform the way we treat diseases. Recent regulatory approvals, hundreds of new clinical trials, the emergence of CRISPR gene editing, and the effectiveness of mRNA vaccines in dramatic response to the COVID-19 pandemic have converged to create tremendous momentum and expectation. However, challenges with this relatively new class of drugs persist and require specialized knowledge and expertise to overcome. This Review explores shared strategies for developing RNA drug platforms, including layering technologies, addressing common biases and identifying gaps in understanding. It discusses the potential of RNA-based therapeutics to transform medicine, as well as the challenges associated with improving applicability, efficacy and safety profiles. Insights gained from RNA modalities such as antisense oligonucleotides (ASOs) and small interfering RNAs are used to identify important next steps for mRNA and gene editing technologies.
Collapse
Affiliation(s)
- John R Androsavich
- RNA Accelerator, Pfizer Inc, Cambridge, MA, USA.
- Ginkgo Bioworks, Boston, MA, USA.
| |
Collapse
|
5
|
Abstract
Biomedical research has long relied on small-animal studies to elucidate disease process and develop new medical treatments. The introduction of in vivo functional imaging technology, such as PET, has allowed investigators to peer inside their subjects and follow disease progression longitudinally as well as improve understanding of normal biological processes. Recent developments in CRISPR, immuno-PET, and high-resolution in vivo imaging have only increased the importance of small-animal, or preclinical, PET imaging. Other drivers of preclinical PET innovation include new combinations of imaging technologies, such as PET/MR imaging, which require changes to PET hardware.
Collapse
Affiliation(s)
- Adrienne L Lehnert
- Department of Radiology, University of Washington, 1959 Northeast Pacific Street, UW Box 356043, Seattle, WA, USA.
| | - Robert S Miyaoka
- Department of Radiology, University of Washington, 1959 Northeast Pacific Street, UW Box 356043, Seattle, WA, USA
| |
Collapse
|
6
|
Zhang J, Wickizer C, Ding W, Van R, Yang L, Zhu B, Yang J, Wang Y, Wang Y, Xu Y, Zhang C, Shen S, Wang C, Shao Y, Ran C. In vivo three-dimensional brain imaging with chemiluminescence probes in Alzheimer's disease models. Proc Natl Acad Sci U S A 2023; 120:e2310131120. [PMID: 38048460 PMCID: PMC10723133 DOI: 10.1073/pnas.2310131120] [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: 06/15/2023] [Accepted: 11/13/2023] [Indexed: 12/06/2023] Open
Abstract
Optical three-dimensional (3D) molecular imaging is highly desirable for providing precise distribution of the target-of-interest in disease models. However, such 3D imaging is still far from wide applications in biomedical research; 3D brain optical molecular imaging, in particular, has rarely been reported. In this report, we designed chemiluminescence probes with high quantum yields, relatively long emission wavelengths, and high signal-to-noise ratios to fulfill the requirements for 3D brain imaging in vivo. With assistance from density-function theory (DFT) computation, we designed ADLumin-Xs by locking up the rotation of the double bond via fusing the furan ring to the phenyl ring. Our results showed that ADLumin-5 had a high quantum yield of chemiluminescence and could bind to amyloid beta (Aβ). Remarkably, ADLumin-5's radiance intensity in brain areas could reach 4 × 107 photon/s/cm2/sr, which is probably 100-fold higher than most chemiluminescence probes for in vivo imaging. Because of its strong emission, we demonstrated that ADLumin-5 could be used for in vivo 3D brain imaging in transgenic mouse models of Alzheimer's disease.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Carly Wickizer
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK73019
| | - Weihua Ding
- Department of Anesthesia Critical Care and Pain Medicine, MGH Center for Translational Pain Research, Massachusetts General Hospital Harvard Medical School, Boston, MA02114
| | - Richard Van
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK73019
| | - Liuyue Yang
- Department of Anesthesia Critical Care and Pain Medicine, MGH Center for Translational Pain Research, Massachusetts General Hospital Harvard Medical School, Boston, MA02114
| | - Biyue Zhu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Jun Yang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Yanli Wang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Yongle Wang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Yulong Xu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Can Zhang
- Genetics and Aging Research Unit, Department of Neurology, McCance Center for Brain Health Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital Harvard Medical School, Charlestown, MA02129
| | - Shiqian Shen
- Department of Anesthesia Critical Care and Pain Medicine, MGH Center for Translational Pain Research, Massachusetts General Hospital Harvard Medical School, Boston, MA02114
| | - Changning Wang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| | - Yihan Shao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK73019
| | - Chongzhao Ran
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Boston, MA02129
| |
Collapse
|
7
|
Zhang J, Wickizer C, Ding W, Van R, Yang L, Zhu B, Yang J, Zhang C, Shen S, Shao Y, Ran C. In Vivo Three-dimensional Brain Imaging with Chemiluminescence Probes in Alzheimer's Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.02.547411. [PMID: 37461700 PMCID: PMC10350002 DOI: 10.1101/2023.07.02.547411] [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: 07/24/2023]
Abstract
Optical three-dimensional (3D) molecular imaging is highly desirable for providing precise distribution of the target-of-interest in disease models. However, such 3D imaging is still far from wide applications in biomedical research; 3D brain optical molecular imaging, in particular, has rarely been reported. In this report, we designed chemiluminescence probes with high quantum yields (QY), relatively long emission wavelengths, and high signal-to-noise ratios (SNRs) to fulfill the requirements for 3D brain imaging in vivo. With assistance from density-function theory (DFT) computation, we designed ADLumin-Xs by locking up the rotation of the double-bond via fusing the furan ring to the phenyl ring. Our results showed that ADLumin-5 had a high quantum yield of chemiluminescence and could bind to amyloid beta (Aβ). Remarkably, ADLumin-5's radiance intensity in brain areas could reach 4×107 photon/s/cm2/sr, which is probably 100-fold higher than most chemiluminescence probes for in vivo imaging. Because of its strong emission, we demonstrated that ADLumin-5 could be used for in vivo 3D brain imaging in transgenic mouse models of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Jing Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
| | - Carly Wickizer
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Weihua Ding
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Richard Van
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Liuyue Yang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Biyue Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
| | - Jun Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Shiqian Shen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yihan Shao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
| |
Collapse
|
8
|
Zhang Q, Song B, Xu Y, Yang Y, Ji J, Cao W, Lu J, Ding J, Cao H, Chu B, Hong J, Wang H, He Y. In vivo bioluminescence imaging of natural bacteria within deep tissues via ATP-binding cassette sugar transporter. Nat Commun 2023; 14:2331. [PMID: 37087540 PMCID: PMC10122673 DOI: 10.1038/s41467-023-37827-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/03/2023] [Indexed: 04/24/2023] Open
Abstract
Most existing bioluminescence imaging methods can only visualize the location of engineered bacteria in vivo, generally precluding the imaging of natural bacteria. Herein, we leverage bacteria-specific ATP-binding cassette sugar transporters to internalize luciferase and luciferin by hitchhiking them on the unique carbon source of bacteria. Typically, the synthesized bioluminescent probes are made of glucose polymer (GP), luciferase, Cy5 and ICG-modified silicon nanoparticles and their substrates are made of GP and D-luciferin-modified silicon nanoparticles. Compared with bacteria with mutations in transporters, which hardly internalize the probes in vitro (i.e., ~2% of uptake rate), various bacteria could robustly engulf the probes with a high uptake rate of around 50%. Notably, the developed strategy enables ex vivo bioluminescence imaging of human vitreous containing ten species of pathogens collected from patients with bacterial endophthalmitis. By using this platform, we further differentiate bacterial and non-bacterial nephritis and colitis in mice, while their chemiluminescent counterparts are unable to distinguish them.
Collapse
Affiliation(s)
- Qian Zhang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yanan Xu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yunmin Yang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Jian Ji
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Wenjun Cao
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Jianping Lu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Jiali Ding
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Haiting Cao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Jiaxu Hong
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China.
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China.
| |
Collapse
|
9
|
Lim SJ, Choi M, Yun I, Lee S, Chang N, Lee CY. Development of Fluorescent Bacteria with Lux and Riboflavin Genes. Int J Mol Sci 2023; 24:ijms24065096. [PMID: 36982169 PMCID: PMC10049116 DOI: 10.3390/ijms24065096] [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: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/30/2023] Open
Abstract
Lumazine protein from marine luminescent bacteria of Photobacterium species bind with very high affinity to the fluorescent chromophore 6,7-dimethyl-8-ribitylumazine. The light emission of bacterial luminescent systems is used as a sensitive, rapid, and safe assay for an ever-increasing number of biological systems. Plasmid pRFN4, containing the genes encoding riboflavin from the rib operon of Bacillus subtilis, was designed for the overproduction of lumazine. To construct fluorescent bacteria for use as microbial sensors, novel recombinant plasmids (pRFN4-Pp N-lumP and pRFN4-Pp luxLP N-lumP) were constructed by amplifying the DNA encoding the N-lumP gene (luxL) from P. phosphoreum and the promoter region (luxLP) present upstream of the lux operon of the gene by PCR and ligating into the pRFN4-Pp N-lumP plasmid. A new recombinant plasmid, pRFN4-Pp luxLP-N-lumP, was constructed with the expectation that the fluorescence intensity would be further increased when transformed into Escherichia coli. When this plasmid was transformed into E. coli 43R, the fluorescence intensity of transformants was 500 times greater than that of E. coli alone. As a result, the recombinant plasmid in which the gene encoding N-LumP and DNA containing the lux promoter exhibited expression that was so high as to show fluorescence in single E. coli cells. The fluorescent bacterial systems developed in the present study using lux and riboflavin genes can be utilized in the future as biosensors with high sensitivity and rapid analysis times.
Collapse
Affiliation(s)
- Sun-Joo Lim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Miae Choi
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Inseop Yun
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Seulgi Lee
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Ny Chang
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Chan-Yong Lee
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
10
|
Yin W, Li X, Cao Q, Wang H, Zhang B. Erratum: Bioluminescence tomography reconstruction in conjunction with an organ probability map as an anatomical reference: erratum. BIOMEDICAL OPTICS EXPRESS 2023; 14:1243-1244. [PMID: 36950246 PMCID: PMC10026586 DOI: 10.1364/boe.470117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 06/18/2023]
Abstract
[This corrects the article on p. 1275 in vol. 13, PMID: 35414991.].
Collapse
Affiliation(s)
- Wanzhou Yin
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xiang Li
- Department of Radiology, the Second Hospital of Dalian Medial University, Dalian, Liaoning 116023, China
| | - Qian Cao
- Department of Radiology, the Second Hospital of Dalian Medial University, Dalian, Liaoning 116023, China
| | - Hongkai Wang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Bin Zhang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| |
Collapse
|
11
|
Deng Z, Xu X, Iordachita I, Dehghani H, Zhang B, Wong JW, Wang KKH. Mobile bioluminescence tomography-guided system for pre-clinical radiotherapy research. BIOMEDICAL OPTICS EXPRESS 2022; 13:4970-4989. [PMID: 36187243 PMCID: PMC9484421 DOI: 10.1364/boe.460737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 06/16/2023]
Abstract
Due to low imaging contrast, a widely-used cone-beam computed tomography-guided small animal irradiator is less adept at localizing in vivo soft tissue targets. Bioluminescence tomography (BLT), which combines a model of light propagation through tissue with an optimization algorithm, can recover a spatially resolved tomographic volume for an internal bioluminescent source. We built a novel mobile BLT system for a small animal irradiator to localize soft tissue targets for radiation guidance. In this study, we elaborate its configuration and features that are indispensable for accurate image guidance. Phantom and in vivo validations show the BLT system can localize targets with accuracy within 1 mm. With the optimal choice of threshold and margin for target volume, BLT can provide a distinctive opportunity for investigators to perform conformal biology-guided irradiation to malignancy.
Collapse
Affiliation(s)
- Zijian Deng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Biomedical Imaging and Radiation Technology Laboratory (BIRTLab), Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- These authors contributed equally to this work
| | - Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Biomedical Imaging and Radiation Technology Laboratory (BIRTLab), Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- These authors contributed equally to this work
| | - Iulian Iordachita
- Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Hamid Dehghani
- School of Computer Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Bin Zhang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - John W Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Biomedical Imaging and Radiation Technology Laboratory (BIRTLab), Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
12
|
Makela AV, Schott MA, Sehl OC, Gevaert JJ, Foster PJ, Contag CH. Tracking the fates of iron-labeled tumor cells in vivo using magnetic particle imaging. NANOSCALE ADVANCES 2022; 4:3617-3623. [PMID: 36134351 PMCID: PMC9400501 DOI: 10.1039/d2na00008c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 07/20/2022] [Indexed: 06/16/2023]
Abstract
The use of imaging to detect and monitor the movement and accumulation of cells in living subjects can provide significant insights that can improve our understanding of metastasis and guide therapeutic development. For cell tracking using Magnetic Resonance Imaging (MRI), cells are labeled with iron oxides and the effects of the iron on water provides contrast. However, due to low specificity and difficulties in quantification with MRI, other modalities and approaches need to be developed. Magnetic Particle Imaging (MPI) is an emerging imaging technique which directly detects iron, allowing for a specific, quantitative and sensitive readout. Here, we use MPI to image iron-labeled tumor cells longitudinally, from implantation and growth at a primary site to movement to distant anatomic sites. In vivo bioluminescent imaging (BLI) was used to localize tumor metastases and computed tomography (CT) allowed for correlation of these signals to anatomic locations. These three imaging modalities provide information on immune escape and metastasis of iron-labeled, and unlabeled, tumor cells, and the accumulation of cell-free iron contrast over time. We localized iron signals by MPI and tumor cells via BLI, and correlated these positive contrast images with CT scans to reveal the anatomic sites with cancer cells; histologic analysis confirmed the presence of iron-labeled tumor cells in the tissues, suggesting that the metastatic cells retained enough iron for MPI detection. The use of multi-modality cell tracking reveals the movement, accumulation and fates of labeled cells that will be helpful understanding cancer progression and guiding the development of targeted therapies.
Collapse
Affiliation(s)
- Ashley V Makela
- Michigan State University, Institute for Quantitative Health Science and Engineering East Lansing MI USA
| | - Melissa A Schott
- Michigan State University, Institute for Quantitative Health Science and Engineering East Lansing MI USA
| | - Olivia C Sehl
- Western University, Robarts Research Institute, Department of Medical Biophysics London ON Canada
| | - Julia J Gevaert
- Western University, Robarts Research Institute, Department of Medical Biophysics London ON Canada
| | - Paula J Foster
- Western University, Robarts Research Institute, Department of Medical Biophysics London ON Canada
| | - Christopher H Contag
- Michigan State University, Institute for Quantitative Health Science and Engineering East Lansing MI USA
- Department of Biomedical Engineering, MSU East Lansing MI USA
| |
Collapse
|
13
|
Duan C, Montgomery MK, Chen X, Ullas S, Stansfield J, McElhanon K, Hirenallur-Shanthappa D. Fully Automated Mouse Echocardiography Analysis Using Deep Convolutional Neural Networks. Am J Physiol Heart Circ Physiol 2022; 323:H628-H639. [PMID: 35984765 DOI: 10.1152/ajpheart.00208.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Echocardiography (echo) is a translationally relevant ultrasound imaging modality widely used to assess cardiac structure and function in preclinical models of heart failure (HF) during research and drug development. Though echo is a very valuable tool, the image analysis is a time consuming, resource demanding process, and is susceptible to inter-reader variability. Recent advancements in deep learning have enabled researchers to automate image processing and reduce analysis time and inter-reader variability in the field of medical imaging. In the present study, we developed a fully automated tool - Mouse Echo Neural Net (MENN) - for the analysis of both long axis brightness (B)-mode and short axis motion (M)-mode images of the left ventricle. MENN is a series of fully convolutional neural networks that were trained and validated using manually segmented B-mode and M-mode echo images of the left ventricle. The segmented images were then used to compute cardiac structural and functional metrics. The performance of MENN was further validated in two preclinical models of HF. MENN achieved excellent correlations (Pearson's r = 0.85 to 0.99) and good to excellent agreement between automated and manual analyses. Further inter-reader variability analysis showed that MENN has better agreements with an expert analyst than both a trained analyst and a novice. Notably, the use of MENN reduced manual analysis time by >92%. In conclusion, we developed an automated echocardiography analysis tool that allows for fast and accurate analysis of B-mode and M-mode mouse echo data and mitigates the issue of inter-reader variability in manual analysis.
Collapse
Affiliation(s)
- Chong Duan
- Early Clinical Development, Pfizer Inc., Cambridge, MA, United States
| | | | - Xian Chen
- Comparative Medicine, Pfizer Inc., Cambridge, MA, United States
| | - Soumya Ullas
- Comparative Medicine, Pfizer Inc., Cambridge, MA, United States
| | - John Stansfield
- Early Clinical Development, Pfizer Inc., Cambridge, MA, United States
| | - Kevin McElhanon
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, United States
| | | |
Collapse
|
14
|
Yin W, Li X, Cao Q, Wang H, Zhang B. Bioluminescence tomography reconstruction in conjunction with an organ probability map as an anatomical reference. BIOMEDICAL OPTICS EXPRESS 2022; 13:1275-1291. [PMID: 35414991 PMCID: PMC8973175 DOI: 10.1364/boe.448862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/15/2022] [Accepted: 01/23/2022] [Indexed: 06/14/2023]
Abstract
To alleviate the ill-posedness of bioluminescence tomography (BLT) reconstruction, anatomical information from computed tomography (CT) or magnetic resonance imaging (MRI) is usually adopted to improve the reconstruction quality. With the anatomical information, different organs could be segmented and assigned with appropriate optical parameters, and the reconstruction could be confined into certain organs. However, image segmentation is a time-consuming and challenging work, especially for the low-contrast organs. In this paper, we present a BLT reconstruction method in conjunction with an organ probability map to effectively incorporate the anatomical information. Instead of using a segmentation with a fixed organ map, an organ probability map is established by registering the CT image of the mouse to the statistical mouse atlas with the constraints of the mouse surface and high-contrast organs (bone and lung). Then the organ probability map of the low-contrast organs, such as the liver and kidney, is determined automatically. After discretization of the mouse torso, a heterogeneous model is established as the input for reconstruction, in which the optical parameter of each node is calculated according to the organ probability map. To take the advantage of the sparse Bayesian Learning (SBL) method in recovering block sparse signals in inverse problems, which is common in BLT applications where the target distribution has the characteristic of sparsity and block structure, a two-step method in conjunction with the organ probability map is presented. In the first step, a fast sparse algorithm, L1-LS, is used to reveal the source distribution on the organ level. In the second step, the bioluminescent source is reconstructed on the pixel level based on the SBL method. Both simulation and in vivo experiments are conducted, and the results demonstrate that the organ probability map in conjunction with the proposed two-step BLT reconstruction method is feasible to accurately reconstruct the localization of the bioluminescent light source.
Collapse
Affiliation(s)
- Wanzhou Yin
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
- Contributed equally
| | - Xiang Li
- Department of Radiology, the Second Hospital of Dalian Medial University, Dalian, Liaoning 116023, China
- Contributed equally
| | - Qian Cao
- Department of Radiology, the Second Hospital of Dalian Medial University, Dalian, Liaoning 116023, China
- Contributed equally
| | - Hongkai Wang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Bin Zhang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| |
Collapse
|
15
|
Yang Y, Zhang M, Zhang W, Chen Y, Zhang T, Chen S, Yuan Y, Liang G, Zhang S. Sensitive sensing of alkaline phosphatase and γ-glutamyltranspeptidase activity for tumor imaging. Analyst 2022; 147:1544-1550. [DOI: 10.1039/d2an00163b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mechanism of bioluminescence phenomenon of the probe P-Bz-Luc in the presence of ALP or GGT.
Collapse
Affiliation(s)
- Yanyun Yang
- College of Chemistry, Zhengzhou University, 100 Kexue Road, Zhengzhou, Henan 450001, China
| | - Miaomiao Zhang
- College of Chemistry, Zhengzhou University, 100 Kexue Road, Zhengzhou, Henan 450001, China
| | - Wenting Zhang
- School of Ecology and Environment, Zhengzhou University, 100 Kexue Road, Zhengzhou, Henan 450001, China
| | - Yinglu Chen
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Tong Zhang
- School of Life Sciences, University of Science and Technology of China, Huangshan Road, Hefei, Anhui 230027, China
| | - Sheng Chen
- College of Chemistry, Zhengzhou University, 100 Kexue Road, Zhengzhou, Henan 450001, China
| | - Yue Yuan
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Gaolin Liang
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Shusheng Zhang
- College of Chemistry, Zhengzhou University, 100 Kexue Road, Zhengzhou, Henan 450001, China
| |
Collapse
|
16
|
Raikwar SP, Thangavel R, Ahmed ME, Selvakumar GP, Kempuraj D, Wu K, Khan O, Bazley K, Bussinger B, Kukulka K, Zaheer S, Iyer SS, Govindarajan R, Burton C, James D, Zaheer A. Real-Time Noninvasive Bioluminescence, Ultrasound and Photoacoustic Imaging in NFκB-RE-Luc Transgenic Mice Reveal Glia Maturation Factor-Mediated Immediate and Sustained Spatio-Temporal Activation of NFκB Signaling Post-Traumatic Brain Injury in a Gender-Specific Manner. Cell Mol Neurobiol 2021; 41:1687-1706. [PMID: 32785863 PMCID: PMC8188847 DOI: 10.1007/s10571-020-00937-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Neurotrauma especially traumatic brain injury (TBI) is the leading cause of death and disability worldwide. To improve upon the early diagnosis and develop precision-targeted therapies for TBI, it is critical to understand the underlying molecular mechanisms and signaling pathways. The transcription factor, nuclear factor kappa B (NFκB), which is ubiquitously expressed, plays a crucial role in the normal cell survival, proliferation, differentiation, function, as well as in disease states like neuroinflammation and neurodegeneration. Here, we hypothesized that real-time noninvasive bioluminescence molecular imaging allows rapid and precise monitoring of TBI-induced immediate and rapid spatio-temporal activation of NFκB signaling pathway in response to Glia maturation factor (GMF) upregulation which in turn leads to neuroinflammation and neurodegeneration post-TBI. To test and validate our hypothesis and to gain novel mechanistic insights, we subjected NFκB-RE-Luc transgenic male and female mice to TBI and performed real-time noninvasive bioluminescence imaging (BLI) as well as photoacoustic and ultrasound imaging (PAI). Our BLI data revealed that TBI leads to an immediate and sustained activation of NFκB signaling. Further, our BLI data suggest that especially in male NFκB-RE-Luc transgenic mice subjected to TBI, in addition to brain, there is widespread activation of NFκB signaling in multiple organs. However, in the case of the female NFκB-RE-Luc transgenic mice, TBI induces a very specific and localized activation of NFκB signaling in the brain. Further, our microRNA data suggest that TBI induces significant upregulation of mir-9-5p, mir-21a-5p, mir-34a-5p, mir-16-3p, as well as mir-155-5p within 24 h and these microRNAs can be successfully used as TBI-specific biomarkers. To the best of our knowledge, this is one of the first and unique study of its kind to report immediate and sustained activation of NFκB signaling post-TBI in a gender-specific manner by utilizing real-time non-invasive BLI and PAI in NFκB-RE-Luc transgenic mice. Our study will prove immensely beneficial to gain novel mechanistic insights underlying TBI, unravel novel therapeutic targets, as well as enable us to monitor in real-time the response to innovative TBI-specific precision-targeted gene and stem cell-based precision medicine.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA.
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Shankar S Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Raghav Govindarajan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA.
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| |
Collapse
|
17
|
Xu X, Deng Z, Dehghani H, Iordachita I, Lim M, Wong JW, Wang KKH. Quantitative Bioluminescence Tomography-guided Conformal Irradiation for Preclinical Radiation Research. Int J Radiat Oncol Biol Phys 2021; 111:1310-1321. [PMID: 34411639 PMCID: PMC8602741 DOI: 10.1016/j.ijrobp.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 10/31/2022]
Abstract
PURPOSE Widely used cone beam computed tomography (CBCT)-guided irradiators in preclinical radiation research are limited to localize soft tissue target because of low imaging contrast. Knowledge of target volume is a fundamental need for radiation therapy (RT). Without such information to guide radiation, normal tissue can be overirradiated, introducing experimental uncertainties. This led us to develop high-contrast quantitative bioluminescence tomography (QBLT) for guidance. The use of a 3-dimensional bioluminescence signal, related to cell viability, for preclinical radiation research is one step toward biology-guided RT. METHODS AND MATERIALS Our QBLT system enables multiprojection and multispectral bioluminescence imaging to maximize input data for the tomographic reconstruction. Accurate quantification of spectrum and dynamic change of in vivo signal were also accounted for the QBLT. A spectral-derivative method was implemented to eliminate the modeling of the light propagation from animal surface to detector. We demonstrated the QBLT capability of guiding conformal RT using a bioluminescent glioblastoma (GBM) model in vivo. A threshold was determined to delineate QBLT reconstructed gross target volume (GTVQBLT), which provides the best overlap between the GTVQBLT and CBCT contrast labeled GBM (GTV), used as the ground truth for GBM volume. To account for the uncertainty of GTVQBLT in target positioning and volume delineation, a margin was determined and added to the GTVQBLT to form a QBLT planning target volume (PTVQBLT) for guidance. RESULTS The QBLT can reconstruct in vivo GBM with localization accuracy within 1 mm. A 0.5-mm margin was determined and added to GTVQBLT to form PTVQBLT, largely improving tumor coverage from 75.0% (0 mm margin) to 97.9% in average, while minimizing normal tissue toxicity. With the goal of prescribed dose 5 Gy covering 95% of PTVQBLT, QBLT-guided 7-field conformal RT can effectively irradiate 99.4 ± 1.0% of GTV. CONCLUSIONS The QBLT provides a unique opportunity for investigators to use biologic information for target delineation, guiding conformal irradiation, and reducing normal tissue involvement, which is expected to increase reproducibility of scientific discovery.
Collapse
Affiliation(s)
- Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zijian Deng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hamid Dehghani
- School of Computer Science, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Iulian Iordachita
- Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Department of Neurosurgery, Stanford University, Stanford, California
| | - John W Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
18
|
Lim S, Oh E, Choi M, Lee E, Lee CY. Generation of Fluorescent Bacteria with the Genes Coding for Lumazine Protein and Riboflavin Biosynthesis. SENSORS 2021; 21:s21134506. [PMID: 34209387 PMCID: PMC8272222 DOI: 10.3390/s21134506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022]
Abstract
Lumazine protein is a member of the riboflavin synthase superfamily and the intense fluorescence is caused by non-covalently bound to 6,7-dimethyl 8-ribityllumazine. The pRFN4 plasmid, which contains the riboflavin synthesis genes from Bacillus subtilis, was originally designed for overproduction of the fluorescent ligand of 6,7-dimethyl 8-ribityllumazine. To provide the basis for a biosensor based on the lux gene from bioluminescent bacteria of Photobacterium leiognathi, the gene coding for N-terminal domain half of the lumazine protein extending to amino acid 112 (N-LumP) and the gene for whole lumazine protein (W-LumP) from P. leiognathi were introduced by polymerase chain reaction (PCR) and ligated into pRFN4 vector, to construct the recombinant plasmids of N-lumP-pRFN4 and W-lumP-pRFN4 as well as their modified plasmids by insertion of the lux promoter. The expression of the genes in the recombinant plasmids was checked in various Escherichia coli strains, and the fluorescence intensity in Escherichia coli 43R can even be observed in a single cell. These results concerning the co-expression of the genes coding for lumazine protein and for riboflavin synthesis raise the possibility to generate fluorescent bacteria which can be used in the field of bio-imaging.
Collapse
|
19
|
Michaels SA, Hennessey KM, Paragas N, Paredez AR, Ojo KK. A Curious Case for Development of Kinase Inhibitors as Antigiardiasis Treatments Using Advanced Drug Techniques. ACS Infect Dis 2021; 7:943-947. [PMID: 33534539 DOI: 10.1021/acsinfecdis.0c00919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Giardiasis is a neglected parasitic diarrheal disease that is particularly associated with poverty. Current treatment options are limited in the face of growing resistance, but the reduced kinome of Giardia lamblia increases the likelihood of identifying nonredundant essential kinases as potential drug targets. Repurposing known and newly identified kinase inhibitors in drug development programs for novel giardiasis therapeutics could therefore be a cost-effective and time saving approach. Innovative improvements to physiologically-based pharmacokinetic modeling coupled with emerging imaging technologies and a CRISPR-interference method could accelerate progress toward the goal of more effective giardiasis therapeutics based on kinase inhibition.
Collapse
Affiliation(s)
- Samantha A Michaels
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Kelly M Hennessey
- Department of Biology, University of Washington, Seattle, Washington 98195, United States
| | - Neal Paragas
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Alexander R Paredez
- Department of Biology, University of Washington, Seattle, Washington 98195, United States
| | - Kayode K Ojo
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| |
Collapse
|
20
|
Emerging tools for bioluminescence imaging. Curr Opin Chem Biol 2021; 63:86-94. [PMID: 33770744 DOI: 10.1016/j.cbpa.2021.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023]
Abstract
Bioluminescence (BL) relies on the enzymatic reaction between luciferase, a substrate conventionally named luciferin, and various cofactors. BL imaging has become a widely used technique to interrogate gene expression and cell fate, both in small and large animal models of research. Recent developments include the generation of improved luciferase-luciferin systems for deeper and more sensitive imaging as well as new caged luciferins to report on enzymatic activity and other intracellular functions. Here, we critically evaluate the emerging tools for BL imaging aiming to provide the reader with an updated compendium of the latest developments (2018-2020) and their notable applications.
Collapse
|
21
|
Ko RB, Soto LA, von Eyben R, Melemenidis S, Rankin EB, Maxim PG, Graves EE, Loo BW. Evaluating the Reproducibility of Mouse Anatomy under Rotation in a Custom Immobilization Device for Conformal FLASH Radiotherapy. Radiat Res 2021; 194:600-606. [PMID: 32857849 DOI: 10.1667/rade-20-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/18/2020] [Indexed: 11/03/2022]
Abstract
The observation of an enhanced therapeutic index for FLASH radiotherapy in mice has created interest in practical laboratory-based FLASH irradiators. To date, systems capable of 3D conformal FLASH irradiation in mice have been lacking. We are developing such a system, incorporating a high-current linear accelerator to produce a collimated X-ray beam in a stationary beamline design, rotating the mouse about a longitudinal axis to achieve conformal irradiation from multiple beam directions. The purpose of this work was to evaluate the reproducibility of mouse anatomy under rotation at speeds compatible with conformal FLASH delivery. Three short-hair mice and two hairless mice were immobilized under anesthesia in body weight-specific contoured plastic molds, and subjected to three rotational (up to 3 revolutions/s) and two non-rotational movement interventions. MicroCT images were acquired before and after each intervention. The displacements of 11 anatomic landmarks were measured on the image pairs. The displacement of the anatomical landmarks with any of the interventions was 0.5 mm or less for 92.4% of measurements, with a single measurement out of 275 (11 landmarks × 5 interventions × 5 mice) reaching 1 mm. There was no significant difference in the displacements associated with rotation compared to those associated with moving the immobilized mouse in and out of a scanner or with leaving the mouse in place for 5 min with no motion. There were no significant differences in displacements between mice with or without hair, although the analysis is limited by small numbers, or between different anatomic landmarks. These results show that anatomic reproducibility under rotation speed corresponding to FLASH irradiation times appears to be compatible with conformal/stereotactic irradiation in mice.
Collapse
Affiliation(s)
- Ryan B Ko
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Luis A Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Peter G Maxim
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
22
|
Functional Imaging Using Bioluminescent Reporter Genes in Living Subjects. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
23
|
Corbett DC, Fabyan WB, Grigoryan B, O'Connor CE, Johansson F, Batalov I, Regier MC, DeForest CA, Miller JS, Stevens KR. Thermofluidic heat exchangers for actuation of transcription in artificial tissues. SCIENCE ADVANCES 2020; 6:6/40/eabb9062. [PMID: 32998880 PMCID: PMC7527231 DOI: 10.1126/sciadv.abb9062] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/13/2020] [Indexed: 05/12/2023]
Abstract
Spatial patterns of gene expression in living organisms orchestrate cell decisions in development, homeostasis, and disease. However, most methods for reconstructing gene patterning in 3D cell culture and artificial tissues are restricted by patterning depth and scale. We introduce a depth- and scale-flexible method to direct volumetric gene expression patterning in 3D artificial tissues, which we call "heat exchangers for actuation of transcription" (HEAT). This approach leverages fluid-based heat transfer from printed networks in the tissues to activate heat-inducible transgenes expressed by embedded cells. We show that gene expression patterning can be tuned both spatially and dynamically by varying channel network architecture, fluid temperature, fluid flow direction, and stimulation timing in a user-defined manner and maintained in vivo. We apply this approach to activate the 3D positional expression of Wnt ligands and Wnt/β-catenin pathway regulators, which are major regulators of development, homeostasis, regeneration, and cancer throughout the animal kingdom.
Collapse
Affiliation(s)
- Daniel C Corbett
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Wesley B Fabyan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Bagrat Grigoryan
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Colleen E O'Connor
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Fredrik Johansson
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Ivan Batalov
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Mary C Regier
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Jordan S Miller
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Cao X, Li K, Xu XL, Deneen KMV, Geng GH, Chen XL. Development of tomographic reconstruction for three-dimensional optical imaging: From the inversion of light propagation to artificial intelligence. Artif Intell Med Imaging 2020; 1:78-86. [DOI: 10.35711/aimi.v1.i2.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/01/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
Optical molecular tomography (OMT) is an imaging modality which uses an optical signal, especially near-infrared light, to reconstruct the three-dimensional information of the light source in biological tissue. With the advantages of being low-cost, noninvasive and having high sensitivity, OMT has been applied in preclinical and clinical research. However, due to its serious ill-posedness and ill-condition, the solution of OMT requires heavy data analysis and the reconstruction quality is limited. Recently, the artificial intelligence (commonly known as AI)-based methods have been proposed to provide a different tool to solve the OMT problem. In this paper, we review the progress on OMT algorithms, from conventional methods to AI-based methods, and we also give a prospective towards future developments in this domain.
Collapse
Affiliation(s)
- Xin Cao
- School of Information Science and Technology, Northwest University, Xi’an 710069, Shaanxi Province, China
| | - Kang Li
- School of Information Science and Technology, Northwest University, Xi’an 710069, Shaanxi Province, China
| | - Xue-Li Xu
- School of Information Science and Technology, Northwest University, Xi’an 710069, Shaanxi Province, China
| | - Karen M von Deneen
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, and School of Life Science and Technology, Xidian University, Xi’an 710126, Shaanxi Province, China
| | - Guo-Hua Geng
- School of Information Science and Technology, Northwest University, Xi’an 710069, Shaanxi Province, China
| | - Xue-Li Chen
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, and School of Life Science and Technology, Xidian University, Xi’an 710126, Shaanxi Province, China
| |
Collapse
|
25
|
Miyaoka RS, Lehnert A. Small animal PET: a review of what we have done and where we are going. Phys Med Biol 2020; 65. [PMID: 32357344 DOI: 10.1088/1361-6560/ab8f71] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
Small animal research is an essential tool in studying both pharmaceutical biodistributions and disease progression over time. Furthermore, through the rapid development of in vivo imaging technology over the last few decades, small animal imaging (also referred to as preclinical imaging) has become a mainstay for all fields of biologic research and a center point for most preclinical cancer research. Preclinical imaging modalities include optical, MRI and MRS, microCT, small animal PET, ultrasound, and photoacoustic, each with their individual strengths. The strong points of small animal PET are its translatability to the clinic; its quantitative imaging capabilities; its whole-body imaging ability to dynamically trace functional/biochemical processes; its ability to provide useful images with only nano- to pico‑ molar concentrations of administered compounds; and its ability to study animals serially over time. This review paper gives an overview of the development and evolution of small animal PET imaging. It provides an overview of detector designs; system configurations; multimodality PET imaging systems; image reconstruction and analysis tools; and an overview of research and commercially available small animal PET systems. It concludes with a look toward developing technologies/methodologies that will further enhance the impact of small animal PET imaging on medical research in the future.
Collapse
Affiliation(s)
- Robert S Miyaoka
- Radiology, University of Washington, Seattle, Washington, UNITED STATES
| | - Adrienne Lehnert
- Radiology, University of Washington, Seattle, Washington, UNITED STATES
| |
Collapse
|
26
|
Development of an embedded multimodality imaging platform for onco-pharmacology using a smart anticancer prodrug as an example. Sci Rep 2020; 10:2661. [PMID: 32060400 PMCID: PMC7021674 DOI: 10.1038/s41598-020-59561-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/28/2020] [Indexed: 11/08/2022] Open
Abstract
Increasingly, in vivo imaging holds a strategic position in bio-pharmaceutical innovation. We will present the implementation of an integrated multimodal imaging setup enabling the assessment of multiple, complementary parameters. The system allows the fusion of information provided by: Near infrared fluorescent biomarkers, bioluminescence (for tumor proliferation status), Photoacoustic and Ultrasound imaging. We will study representative applications to the development of a smart prodrug, delivering a highly cytotoxic chemotherapeutic agent to cancer tumors. The results realized the ability of this embedded, multimodality imaging platform to firstly detect bioluminescent and fluorescent signals, and secondly, record ultrasound and photoacoustic data from the same animal. This study demonstrated that the prodrug was effective in three different models of hypoxia in human cancers compared to the parental cytotoxic agent and the vehicle groups. Monitoring by photoacoustic imaging during the treatments revealed that the prodrug exhibits an intrinsic capability to prevent the progression of tumor hypoxia. It is essential for onco-pharmacology studies to precisely document the hypoxic status of tumors both before and during the time course of treatments. This approach opens new perspectives for exploitation of preclinical mouse models of cancer, especially when considering associations between hypoxia, neoangiogenesis and antitumor activity.
Collapse
|
27
|
Wang H, Han Y, Chen Z, Hu R, Chatziioannou AF, Zhang B. Prediction of major torso organs in low-contrast micro-CT images of mice using a two-stage deeply supervised fully convolutional network. Phys Med Biol 2019; 64:245014. [PMID: 31747654 DOI: 10.1088/1361-6560/ab59a4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Delineation of major torso organs is a key step of mouse micro-CT image analysis. This task is challenging due to low soft tissue contrast and high image noise, therefore anatomical prior knowledge is needed for accurate prediction of organ regions. In this work, we develop a deeply supervised fully convolutional network which uses the organ anatomy prior learned from independently acquired contrast-enhanced micro-CT images to assist the segmentation of non-enhanced images. The network is designed with a two-stage workflow which firstly predicts the rough regions of multiple organs and then refines the accuracy of each organ in local regions. The network is trained and evaluated with 40 mouse micro-CT images. The volumetric prediction accuracy (Dice score) varies from 0.57 for the spleen to 0.95 for the heart. Compared to a conventional atlas registration method, our method dramatically improves the Dice of the abdominal organs by 18%-26%. Moreover, the incorporation of anatomical prior leads to more accurate results for small-sized low-contrast organs (e.g. the spleen and kidneys). We also find that the localized stage of the network has better accuracy than the global stage, indicating that localized single organ prediction is more accurate than global multiple organ prediction. With this work, the accuracy and efficiency of mouse micro-CT image analysis are greatly improved and the need for using contrast agent and high x-ray dose is potentially reduced.
Collapse
Affiliation(s)
- Hongkai Wang
- School of Biomedical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, People's Republic of China
| | | | | | | | | | | |
Collapse
|
28
|
Deng Z, Xu X, Garzon-Muvdi T, Xia Y, Kim E, Belcaid Z, Luksik A, Maxwell R, Choi J, Wang H, Yu J, Iordachita I, Lim M, Wong JW, Wang KKH. In Vivo Bioluminescence Tomography Center of Mass-Guided Conformal Irradiation. Int J Radiat Oncol Biol Phys 2019; 106:612-620. [PMID: 31738948 DOI: 10.1016/j.ijrobp.2019.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 01/21/2023]
Abstract
PURPOSE The cone-beam computed tomography (CBCT)-guided small animal radiation research platform (SARRP) has provided unique opportunities to test radiobiologic hypotheses. However, CBCT is less adept to localize soft tissue targets growing in a low imaging contrast environment. Three-dimensional bioluminescence tomography (BLT) provides strong image contrast and thus offers an attractive solution. We introduced a novel and efficient BLT-guided conformal radiation therapy and demonstrated it in an orthotopic glioblastoma (GBM) model. METHODS AND MATERIALS A multispectral BLT system was integrated with SARRP for radiation therapy (RT) guidance. GBM growth curve was first established by contrast CBCT/magnetic resonance imaging (MRI) to derive equivalent sphere as approximated gross target volume (aGTV). For BLT, mice were subject to multispectral bioluminescence imaging, followed by SARRP CBCT imaging and optical reconstruction. The CBCT image was acquired to generate anatomic mesh for the reconstruction and RT planning. To ensure high accuracy of the BLT-reconstructed center of mass (CoM) for target localization, we optimized the optical absorption coefficients μa by minimizing the distance between the CoMs of BLT reconstruction and contrast CBCT/MRI-delineated GBM volume. The aGTV combined with the uncertainties of BLT CoM localization and target volume determination was used to generate estimated target volume (ETV). For conformal irradiation procedure, the GBM was first localized by the predetermined ETV centered at BLT-reconstructed CoM, followed by SARRP radiation. The irradiation accuracy was qualitatively confirmed by pathologic staining. RESULTS Deviation between CoMs of BLT reconstruction and contrast CBCT/MRI-imaged GBM is approximately 1 mm. Our derived ETV centered at BLT-reconstructed CoM covers >95% of the tumor volume. Using the second-week GBM as an example, the ETV-based BLT-guided irradiation can cover 95.4% ± 4.7% tumor volume at prescribed dose. The pathologic staining demonstrated the BLT-guided irradiated area overlapped well with the GBM location. CONCLUSIONS The BLT-guided RT enables 3-dimensional conformal radiation for important orthotopic tumor models, which provides investigators a new preclinical research capability.
Collapse
Affiliation(s)
- Zijian Deng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eileen Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew Luksik
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John Choi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hailun Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jingjing Yu
- School of Physics and Information Technology, Shaanxi Normal University, Shanxi, China
| | - Iulian Iordachita
- Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John W Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
29
|
Miyazaki T, Gharib SA, Hsu YWA, Xu K, Khodakivskyi P, Kobayashi A, Paragas J, Klose AD, Francis KP, Dubikovskaya E, Page-McCaw PS, Barasch J, Paragas N. Cell-specific image-guided transcriptomics identifies complex injuries caused by ischemic acute kidney injury in mice. Commun Biol 2019; 2:326. [PMID: 31508501 PMCID: PMC6718519 DOI: 10.1038/s42003-019-0571-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/09/2019] [Indexed: 01/20/2023] Open
Abstract
The kidney's inherent complexity has made identifying cell-specific pathways challenging, particularly when temporally associating them with the dynamic pathophysiology of acute kidney injury (AKI). Here, we combine renal cell-specific luciferase reporter mice using a chemoselective luciferin to guide the acquisition of cell-specific transcriptional changes in C57BL/6 background mice. Hydrogen peroxide generation, a common mechanism of tissue damage, was tracked using a peroxy-caged-luciferin to identify optimum time points for immunoprecipitation of labeled ribosomes for RNA-sequencing. Together, these tools revealed a profound impact of AKI on mitochondrial pathways in the collecting duct. In fact, targeting the mitochondria with an antioxidant, ameliorated not only hydrogen peroxide generation, but also significantly reduced oxidative stress and the expression of the AKI biomarker, LCN2. This integrative approach of coupling physiological imaging with transcriptomics and drug testing revealed how the collecting duct responds to AKI and opens new venues for cell-specific predictive monitoring and treatment.
Collapse
Affiliation(s)
- Tomoaki Miyazaki
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
- 2Division of Nephrology, Department of Medicine, Showa University, Yokohama, Japan
| | - Sina A Gharib
- 3Computational Medicine Core, Center for Lung Biology, University of Washington, Seattle, WA 98195 USA
| | - Yun-Wei A Hsu
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | - Katherine Xu
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Pavlo Khodakivskyi
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Akio Kobayashi
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | | | | | | | - Elena Dubikovskaya
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Patrick S Page-McCaw
- 9Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Jonathan Barasch
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Neal Paragas
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|