1
|
Napieczyńska H, Kedziora SM, Haase N, Müller DN, Heuser A, Dechend R, Kräker K. μCT imaging of a multi-organ vascular fingerprint in rats. PLoS One 2024; 19:e0308601. [PMID: 39401231 PMCID: PMC11472947 DOI: 10.1371/journal.pone.0308601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/28/2024] [Indexed: 10/17/2024] Open
Abstract
The importance of microvascular imaging in diagnosis and therapeutic targeting of various diseases is increasingly recognized. The new approach emphasizes the need for holistic studies to understand the inter-organ vascular cross-talk. Here, we report on the development of a novel perfusion protocol which consistently delivers a micro-computed tomography contrast agent to micro-vessels of multiple organs in a single experimental animal. We describe the achieved repeatability of the perfusions, as well as the image analysis steps developed individually for each organ type. We also optimize image acquisition by investigating the compromise between shortening of the scanning time and preservation of the highest possible spatial resolution. Taking together, with the multi-organ perfusion, optimized image acquisition, and the conceived image analysis steps, we provide a comprehensive and reliable experimental protocol for studying vascular morphology and pathology in multi-organ diseases.
Collapse
Affiliation(s)
- Hanna Napieczyńska
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sarah M. Kedziora
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center–A Joint Cooperation between the Max Delbrück Center for Molecular Medicine and the Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadine Haase
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center–A Joint Cooperation between the Max Delbrück Center for Molecular Medicine and the Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Dominik N. Müller
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center–A Joint Cooperation between the Max Delbrück Center for Molecular Medicine and the Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany
| | - Arnd Heuser
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ralf Dechend
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center–A Joint Cooperation between the Max Delbrück Center for Molecular Medicine and the Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany
| | - Kristin Kräker
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center–A Joint Cooperation between the Max Delbrück Center for Molecular Medicine and the Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
2
|
Goossens E, Deblock L, Caboor L, Eynden DVD, Josipovic I, Isaacura PR, Maksimova E, Van Impe M, Bonnin A, Segers P, Cornillie P, Boone MN, Van Driessche I, De Spiegelaere W, De Roo J, Sips P, De Buysser K. From Corrosion Casting to Virtual Dissection: Contrast-Enhanced Vascular Imaging using Hafnium Oxide Nanocrystals. SMALL METHODS 2024; 8:e2301499. [PMID: 38200600 DOI: 10.1002/smtd.202301499] [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: 11/28/2023] [Indexed: 01/12/2024]
Abstract
Vascular corrosion casting is a method used to visualize the three dimensional (3D) anatomy and branching pattern of blood vessels. A polymer resin is injected in the vascular system and, after curing, the surrounding tissue is removed. The latter often deforms or even fractures the fragile cast. Here, a method is proposed that does not require corrosion, and is based on in situ micro computed tomography (micro-CT) scans. To overcome the lack of CT contrast between the polymer cast and the animals' surrounding soft tissue, hafnium oxide nanocrystals (HfO2 NCs) are introduced as CT contrast agents into the resin. The NCs dramatically improve the overall CT contrast of the cast and allow for straightforward segmentation in the CT scans. Careful design of the NC surface chemistry ensures the colloidal stability of the NCs in the casting resin. Using only 5 m% of HfO2 NCs, high-quality cardiovascular casts of both zebrafish and mice can be automatically segmented using CT imaging software. This allows to differentiate even μ $\umu$ m-scale details without having to alter the current resin injection methods. This new method of virtual dissection by visualizing casts in situ using contrast-enhanced CT imaging greatly expands the application potential of the technique.
Collapse
Affiliation(s)
- Eline Goossens
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Loren Deblock
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
| | - Lisa Caboor
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | - Dietger Van den Eynden
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Iván Josipovic
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | - Pablo Reyes Isaacura
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
- Centre for Polymer Material Technologies, Ghent University, Ghent, 9052, Belgium
- Laboratory for Chemical Technology, Ghent University, Ghent, 9052, Belgium
| | - Elizaveta Maksimova
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
- Swiss Nanoscience Institute, University of Basel, Basel, 4056, Switzerland
| | - Matthias Van Impe
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Anne Bonnin
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
| | - Patrick Segers
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Pieter Cornillie
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Matthieu N Boone
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | | | - Ward De Spiegelaere
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Jonathan De Roo
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Patrick Sips
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | | |
Collapse
|
3
|
Jacobs K, Docter D, de Smit L, Korfage HAM, Visser SC, Lobbezoo F, Hlushchuk R, de Bakker BS. High resolution imaging of human development: shedding light on contrast agents. Neuroradiology 2024; 66:1481-1493. [PMID: 38995394 PMCID: PMC11322402 DOI: 10.1007/s00234-024-03413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Visualizing (micro)vascular structures remains challenging for researchers and clinicians due to limitations in traditional radiological imaging methods. Exploring the role of vascular development in craniofacial malformations in experimental settings can enhance understanding of these processes, with the effectiveness of high-resolution imaging techniques being crucial for successful research in this field. Micro-CT imaging offers 3D microstructural insights, but requires contrast-enhancing staining agents (CESAs) for visualizing (micro)-vascular tissues, known as contrast-enhanced micro-CT (CECT). As effective contrast agents are crucial for optimal visualization, this review focuses on comparative studies investigating such agents for micro-vascular tissue imaging using micro-CT. Furthermore, we demonstrate the utilization of B-Lugol solution as a promising contrast agent for acquiring high-quality micro-CT images of (micro)vascular structures in human embryonic samples. METHOD This scoping review followed Preferred Reporting Items for Systematic Reviews and Meta-analysis Protocols. PubMed database provided relevant articles, screened initially by title and abstract. Inclusion and exclusion criteria defined outcomes of interest. RESULTS From an initial search, 273 records were identified, narrowed down to 9 articles after applying our criteria. Additionally, two articles were added through citation searching. This, a total of 11 articles were incorporated in this study. CONCLUSION This micro-CT contrast agent review underscores the need for tailored choices based on research goals. Both Barium sulfate and Iodine-based agents showing excellent results, providing high resolution (micro) vascular content, especially in ex-vivo specimens. However, careful consideration of protocols and tissue characteristics remains imperative for optimizing the effectiveness of micro-CT imaging for the study of cranio-facial vascular development.
Collapse
Affiliation(s)
- Karl Jacobs
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands.
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands.
| | - Daniel Docter
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands
| | - Lotte de Smit
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Hans A M Korfage
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Sophie C Visser
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands
| | - Frank Lobbezoo
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Ruslan Hlushchuk
- Micro-CT Research Group, Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3012, Bern, Switzerland
| | - Bernadette S de Bakker
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam Reproduction & Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Department of Pediatric Surgery, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Phoon CK, Aristizábal O, Farhoud M, Turnbull DH, Wadghiri YZ. Mouse Cardiovascular Imaging. Curr Protoc 2024; 4:e1116. [PMID: 39222027 PMCID: PMC11371386 DOI: 10.1002/cpz1.1116] [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] [Indexed: 09/04/2024]
Abstract
The mouse is the mammalian model of choice for investigating cardiovascular biology, given our ability to manipulate it by genetic, pharmacologic, mechanical, and environmental means. Imaging is an important approach to phenotyping both function and structure of cardiac and vascular components. This review details commonly used imaging approaches, with a focus on echocardiography and magnetic resonance imaging, with brief overviews of other imaging modalities. In this update, we also emphasize the importance of rigor and reproducibility in imaging approaches, experimental design, and documentation. Finally, we briefly outline emerging imaging approaches but caution that reliability and validity data may be lacking. © 2024 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Colin K.L. Phoon
- Division of Pediatric Cardiology, Department of Pediatrics, New York University Grossman School of Medicine, New York, NY
| | - Orlando Aristizábal
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Preclinical Imaging, Division for Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY
| | | | - Daniel H. Turnbull
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Youssef Z. Wadghiri
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Preclinical Imaging, Division for Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
5
|
Nadkarni R, Han ZY, Anderson RJ, Allphin AJ, Clark DP, Badea A, Badea CT. High-resolution hybrid micro-CT imaging pipeline for mouse brain region segmentation and volumetric morphometry. PLoS One 2024; 19:e0303288. [PMID: 38781243 PMCID: PMC11115241 DOI: 10.1371/journal.pone.0303288] [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: 09/28/2023] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Brain region segmentation and morphometry in humanized apolipoprotein E (APOE) mouse models with a human NOS2 background (HN) contribute to Alzheimer's disease (AD) research by demonstrating how various risk factors affect the brain. Photon-counting detector (PCD) micro-CT provides faster scan times than MRI, with superior contrast and spatial resolution to energy-integrating detector (EID) micro-CT. This paper presents a pipeline for mouse brain imaging, segmentation, and morphometry from PCD micro-CT. METHODS We used brains of 26 mice from 3 genotypes (APOE22HN, APOE33HN, APOE44HN). The pipeline included PCD and EID micro-CT scanning, hybrid (PCD and EID) iterative reconstruction, and brain region segmentation using the Small Animal Multivariate Brain Analysis (SAMBA) tool. We applied SAMBA to transfer brain region labels from our new PCD CT atlas to individual PCD brains via diffeomorphic registration. Region-based and voxel-based analyses were used for comparisons by genotype and sex. RESULTS Together, PCD and EID scanning take ~5 hours to produce images with a voxel size of 22 μm, which is faster than MRI protocols for mouse brain morphometry with voxel size above 40 μm. Hybrid iterative reconstruction generates PCD images with minimal artifacts and higher spatial resolution and contrast than EID images. Our PCD atlas is qualitatively and quantitatively similar to the prior MRI atlas and successfully transfers labels to PCD brains in SAMBA. Male and female mice had significant volume differences in 26 regions, including parts of the entorhinal cortex and cingulate cortex. APOE22HN brains were larger than APOE44HN brains in clusters from the hippocampus, a region where atrophy is associated with AD. CONCLUSIONS This work establishes a pipeline for mouse brain analysis using PCD CT, from staining to imaging and labeling brain images. Our results validate the effectiveness of the approach, setting a foundation for research on AD mouse models while reducing scanning durations.
Collapse
Affiliation(s)
- Rohan Nadkarni
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Zay Yar Han
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Robert J. Anderson
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Alex J. Allphin
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Darin P. Clark
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Alexandra Badea
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Cristian T. Badea
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
6
|
Ma Y, Sun W, Bai J, Gao F, Ma H, Liu H, Hu J, Xu C, Zhang X, Liu Z, Yuan T, Sun C, Huang Y, Wang R. Targeting blood brain barrier-Remote ischemic conditioning alleviates cognitive impairment in female APP/PS1 rats. CNS Neurosci Ther 2024; 30:e14613. [PMID: 38379185 PMCID: PMC10879645 DOI: 10.1111/cns.14613] [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: 06/16/2023] [Revised: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a significant global health concern, and it is crucial that we find effective methods to prevent or slow down AD progression. Recent studies have highlighted the essential role of blood vessels in clearing Aβ, a protein that contributes to AD. Scientists are exploring blood biomarkers as a potential tool for future AD diagnosis. One promising method that may help prevent AD is remote ischemic conditioning (RIC). RIC involves using sub-lethal ischemic-reperfusion cycles on limbs. However, a comprehensive understanding of how RIC can prevent AD and its long-term effectiveness is still lacking. Further research is essential to fully comprehend the potential benefits of RIC in preventing AD. METHODS Female wild-type (WT) and APP/PS1 transgenic rats, aged 12 months, underwent ovariectomy and were subsequently assigned to WT, APP/PS1, and APP/PS1 + RIC groups. RIC was conducted five times a week for 4 weeks. The rats' depressive and cognitive behaviors were evaluated using force swimming, open-field tests, novel objective recognition, elevated plus maze, and Barnes maze tests. Evaluation of the neurovascular unit (NVU), synapses, vasculature, astrocytes, and microglia was conducted using immunofluorescence staining (IF), Western blot (WB), and transmission electron microscopy (TEM). Additionally, the cerebro-vasculature was examined using micro-CT, and cerebral blood flow (CBF) was measured using Speckle Doppler. Blood-brain barrier (BBB) permeability was determined by measuring the Evans blue leakage. Finally, Aβ levels in the rat frontal cortex were measured using WB, ELISA, or IF staining. RESULTS RIC enhanced memory-related protein expression and rescued depressive-like behavior and cognitive decline in APP/PS1 transgenic rats. Additionally, the intervention protected NVU in the rat frontal cortex, as evidenced by (1) increased expression of TJ (tight junction) proteins, pericyte marker PDGFRβ, and glucose transporter 1 (GLUT1), as well as decreased VCAM1; (2) mitigation of ultrastructure impairment in neuron, cerebral vascular, and astrocyte; (3) upregulation of A2 astrocyte phenotype markers and downregulation of A1 phenotype markers, indicating a shift toward a healthier phenotype. Correspondingly, RIC intervention alleviated neuroinflammation, as evidenced by the decreased Iba1 level, a microglia marker. Meanwhile, RIC intervention elevated CBF in frontal cortex of the rats. Notably, RIC intervention effectively suppressed Aβ toxicity, as demonstrated by the enhancement of α-secretase and attenuation of β-secretase (BACE1) and γ- secretase and Aβ1-42 and Aβ1-40 levels as well. CONCLUSION Chronic RIC intervention exerts vascular and neuroprotective roles, suggesting that RIC could be a promising therapeutic strategy targeting the BBB and NVU during AD development.
Collapse
Affiliation(s)
- Yuxuan Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Wuxiang Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Jing Bai
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Fujia Gao
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Haoran Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Huiyu Liu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Jiewei Hu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Chao Xu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Xin Zhang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Zixuan Liu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Tao Yuan
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Chenxu Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Yuanyuan Huang
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Ruimin Wang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| |
Collapse
|
7
|
Zdun M, Melnyk OP, Melnyk OO, Nabzdyk M. Blood supply to the cranial cavity in the patagonian mara (Dolichotis patagonum). Vet Res Commun 2023; 47:2199-2205. [PMID: 36976443 DOI: 10.1007/s11259-023-10113-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Rodents are the most numerous order of mammals. The literature presents information on the arterial circle of the brain in capybara, the guinea pig of the family Caviidae and many other not so closely related rodent species. Information on the blood supply to the brain is often incomplete and focuses on one pathway in a broader comparative aspect. The supply of oxygen and nutrients to the brain is very important for its proper functioning. The aim of this study is to describe the pathways supplying blood to the cranial cavity and to describe the arterial circle of the brain in the Patagonian mara. The study was conducted on 46 specimens using two methods. The first of them used a stained solution of the chemo-setting acrylic material. The second one, the colored liquid LBS 3060 latex. The arterial circle of the brain is a heart-shaped structure. It is formed by rostral cerebral arteries, caudal communicating arteries and the basilar artery. Blood supplies the arterial circle of the brain in three ways. First one is the basilar artery, which originates from the vertebral arteries. The second one is the internal carotid artery which joins a branch from the external ophthalmic artery. The third is the internal ophthalmic artery, which branches from the external ophthalmic artery.
Collapse
Affiliation(s)
- Maciej Zdun
- Department of Animal Anatomy, Poznan University of Life Sciences, Wojska Polskiego 71C, Poznań, 60-625, Poland.
- Department of Basic and Preclinical Sciences, Nicolaus Copernicus University, Lwowska 1, Toruń, 87-100, Poland.
- Department of Animal Anatomy, Histology and Pathomorphology, National University of Life and Environmental Sciences of Ukraine, Heroiv Oborony Str.15, Kyiv, 03041, Ukraine.
| | - Oleg P Melnyk
- Department of Animal Anatomy, Histology and Pathomorphology, National University of Life and Environmental Sciences of Ukraine, Heroiv Oborony Str.15, Kyiv, 03041, Ukraine
| | - Oleksii O Melnyk
- Department of Animal Anatomy, Histology and Pathomorphology, National University of Life and Environmental Sciences of Ukraine, Heroiv Oborony Str.15, Kyiv, 03041, Ukraine
| | - Maria Nabzdyk
- Department of Animal Anatomy, Poznan University of Life Sciences, Wojska Polskiego 71C, Poznań, 60-625, Poland
| |
Collapse
|
8
|
Koepple C, Pollmann L, Pollmann NS, Schulte M, Kneser U, Gretz N, Schmidt VJ. Microporous Polylactic Acid Scaffolds Enable Fluorescence-Based Perfusion Imaging of Intrinsic In Vivo Vascularization. Int J Mol Sci 2023; 24:14813. [PMID: 37834261 PMCID: PMC10573679 DOI: 10.3390/ijms241914813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
In vivo tissue engineering (TE) techniques like the AV loop model provide an isolated and well-defined microenvironment to study angiogenesis-related cell interactions. Functional visualization of the microvascular network within these artificial tissue constructs is crucial for the fundamental understanding of vessel network formation and to identify the underlying key regulatory mechanisms. To facilitate microvascular tracking advanced fluorescence imaging techniques are required. We studied the suitability of microporous polylactic acid (PLA) scaffolds with known low autofluorescence to form axial vascularized tissue constructs in the AV loop model and to validate these scaffolds for fluorescence-based perfusion imaging. Compared to commonly used collagen elastin (CE) scaffolds, the total number of vessels and cells in PLA scaffolds was lower. In detail, CE-based constructs exhibited significantly higher vessel numbers on day 14 and 28 (d14: 316 ± 53; d28: 610 ± 74) compared to the respective time points in PLA-based constructs (d14: 144 ± 18; d28: 327 ± 34; each p < 0.05). Analogously, cell counts in CE scaffolds were higher compared to corresponding PLA constructs (d14: 7661.25 ± 505.93 and 5804.04 ± 716.59; d28: 11211.75 + 1278.97 and 6045.71 ± 572.72, p < 0.05). CE scaffolds showed significantly higher vessel densities in proximity to the main vessel axis compared to PLA scaffolds (200-400 µm and 600-800 µm on day 14; 400-1000 µm and 1400-1600 µm on day 28). CE scaffolds had significantly higher cell counts on day 14 at distances from 800 to 2000 µm and at distances from 400 to 1600 µm on day 28. While the total number of vessels and cells in PLA scaffolds were lower, both scaffold types were ideally suited for axial vascularization techniques. The intravascular perfusion of PLA-based constructs with fluorescence dye MHI148-PEI demonstrated dye specificity against vascular walls of low- and high-order branches as well as capillaries and facilitated the fluorescence-based visualization of microcirculatory networks. Fluorophore tracking may contribute to the development of automated quantification methods after 3D reconstruction and image segmentation. These technologies may facilitate the characterization of key regulators within specific subdomains and add to the current understanding of vessel formation in axially vascularized tissue constructs.
Collapse
Affiliation(s)
- Christoph Koepple
- Department for Hand-, Plastic- and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Heidelberg University, 67071 Ludwigshafen, Germany; (L.P.); (N.S.P.); (M.S.); (U.K.)
| | - Lukas Pollmann
- Department for Hand-, Plastic- and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Heidelberg University, 67071 Ludwigshafen, Germany; (L.P.); (N.S.P.); (M.S.); (U.K.)
| | - Nicola Sariye Pollmann
- Department for Hand-, Plastic- and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Heidelberg University, 67071 Ludwigshafen, Germany; (L.P.); (N.S.P.); (M.S.); (U.K.)
| | - Matthias Schulte
- Department for Hand-, Plastic- and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Heidelberg University, 67071 Ludwigshafen, Germany; (L.P.); (N.S.P.); (M.S.); (U.K.)
| | - Ulrich Kneser
- Department for Hand-, Plastic- and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Heidelberg University, 67071 Ludwigshafen, Germany; (L.P.); (N.S.P.); (M.S.); (U.K.)
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
| | - Volker J. Schmidt
- Department of Plastic Surgery and Hand Surgery, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland;
| |
Collapse
|
9
|
Zhang H, Kang DH, Piantino M, Tominaga D, Fujimura T, Nakatani N, Taylor JN, Furihata T, Matsusaki M, Fujita S. Rapid Quantification of Microvessels of Three-Dimensional Blood-Brain Barrier Model Using Optical Coherence Tomography and Deep Learning Algorithm. BIOSENSORS 2023; 13:818. [PMID: 37622905 PMCID: PMC10452445 DOI: 10.3390/bios13080818] [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: 06/19/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
The blood-brain barrier (BBB) is a selective barrier that controls the transport between the blood and neural tissue features and maintains brain homeostasis to protect the central nervous system (CNS). In vitro models can be useful to understand the role of the BBB in disease and assess the effects of drug delivery. Recently, we reported a 3D BBB model with perfusable microvasculature in a Transwell insert. It replicates several key features of the native BBB, as it showed size-selective permeability of different molecular weights of dextran, activity of the P-glycoprotein efflux pump, and functionality of receptor-mediated transcytosis (RMT), which is the most investigated pathway for the transportation of macromolecules through endothelial cells of the BBB. For quality control and permeability evaluation in commercial use, visualization and quantification of the 3D vascular lumen structures is absolutely crucial. Here, for the first time, we report a rapid, non-invasive optical coherence tomography (OCT)-based approach to quantify the microvessel network in the 3D in vitro BBB model. Briefly, we successfully obtained the 3D OCT images of the BBB model and further processed the images using three strategies: morphological imaging processing (MIP), random forest machine learning using the Trainable Weka Segmentation plugin (RF-TWS), and deep learning using pix2pix cGAN. The performance of these methods was evaluated by comparing their output images with manually selected ground truth images. It suggested that deep learning performed well on object identification of OCT images and its computation results of vessel counts and surface areas were close to the ground truth results. This study not only facilitates the permeability evaluation of the BBB model but also offers a rapid, non-invasive observational and quantitative approach for the increasing number of other 3D in vitro models.
Collapse
Affiliation(s)
- Huiting Zhang
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (H.Z.); (J.N.T.); (M.M.)
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (D.-H.K.); (M.P.)
| | - Dong-Hee Kang
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (D.-H.K.); (M.P.)
| | - Marie Piantino
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (D.-H.K.); (M.P.)
| | - Daisuke Tominaga
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba 305-8565, Ibaraki, Japan;
| | - Takashi Fujimura
- SCREEN Holdings Co., Ltd., 322 Furukawa-cho, Hazukashi, Fushimi-ku, Kyoto 612-8486, Kyoto, Japan; (T.F.); (N.N.)
| | - Noriyuki Nakatani
- SCREEN Holdings Co., Ltd., 322 Furukawa-cho, Hazukashi, Fushimi-ku, Kyoto 612-8486, Kyoto, Japan; (T.F.); (N.N.)
| | - J. Nicholas Taylor
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (H.Z.); (J.N.T.); (M.M.)
| | - Tomomi Furihata
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji 192-0392, Tokyo, Japan;
| | - Michiya Matsusaki
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (H.Z.); (J.N.T.); (M.M.)
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (D.-H.K.); (M.P.)
| | - Satoshi Fujita
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (H.Z.); (J.N.T.); (M.M.)
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (D.-H.K.); (M.P.)
| |
Collapse
|
10
|
Schneider B, Kopf KW, Mason E, Dawson M, Coronado Escobar D, Majka SM. Microcomputed tomography visualization and quantitation of the pulmonary arterial microvascular tree in mouse models of chronic lung disease. Pulm Circ 2023; 13:e12279. [PMID: 37645586 PMCID: PMC10461042 DOI: 10.1002/pul2.12279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Pulmonary vascular dysfunction is characterized by remodeling and loss of microvessels in the lung and is a major manifestation of chronic lung diseases (CLD). In murine models of CLD, the small arterioles and capillaries are the first and most prevalent vessels that are affected by pruning and remodeling. Thus, visualization of the pulmonary arterial vasculature in three dimensions is essential to define pruning and remodeling both temporally and spatially and its role in the pathogenesis of CLD, aging, and tissue repair. To this end, we have developed a novel method to visualize and quantitate the murine pulmonary arterial circulation using microcomputed tomography (µCT) imaging. Using this perfusion technique, we can quantitate microvessels to approximately 6 µM in diameter. We hypothesize that bleomycin-induced injury would have a significant impact on the arterial vascular structure. As proof of principle, we demonstrated that as a result of bleomycin-induced injury at peak fibrosis, significant alterations in arterial vessel structure were visible in the three-dimensional models as well as quantification. Thus, we have successfully developed a perfusion methodology and complementary analysis techniques, which allows for the reconstruction, visualization, and quantitation of the mouse pulmonary arterial microvasculature in three-dimensions. This tool will further support the examination and understanding of angiogenesis during the development of CLD as well as repair following injury.
Collapse
Affiliation(s)
- Ben Schneider
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverColoradoUSA
| | - Katrina W. Kopf
- Biological Resource CenterNational Jewish HealthDenverColoradoUSA
| | - Emma Mason
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverColoradoUSA
| | - Maggie Dawson
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverColoradoUSA
| | | | - Susan M. Majka
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Gates Center for Regenerative Medicine and Stem Cell BiologyUniversity of ColoradoAuroraColoradoUSA
| |
Collapse
|
11
|
Zhu J, Liu X, Xu J, Deng Y, Wang P, Liu Z, Yang Q, Li D, Yu T, Zhu D. A versatile vessel casting method for fine mapping of vascular networks using a hydrogel-based lipophilic dye solution. CELL REPORTS METHODS 2023; 3:100407. [PMID: 36936073 PMCID: PMC10014313 DOI: 10.1016/j.crmeth.2023.100407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/11/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Efficient labeling of the vasculature is important for understanding the organization of vascular networks. Here, we propose VALID, a vessel-labeling method that enables visualization of vascular networks with tissue clearing and light-sheet microscopy. VALID transforms traditional lipophilic dye solution into hydrogel by introducing gelatin and restrains the dye aggregation, resulting in complete and uniform vessel-labeling patterns with high signal-to-background ratios. VALID also enhances the compatibility of lipophilic dyes with solvent-based tissue-clearing protocols, which was hard to achieve previously. Using VALID, we combined lipophilic dyes with solvent-based tissue-clearing techniques to perform 3D reconstructions of vasculature within mouse brain and spinal cord. We also employed VALID for 3D visualization and quantification of microvascular damage in a middle cerebral artery occlusion mouse model. VALID should provide a simple, cost-effective vessel-labeling protocol that would significantly widen the applications of lipophilic dyes in research on cerebrovascular complications.
Collapse
Affiliation(s)
- Jingtan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Xiaomei Liu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Jianyi Xu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Yating Deng
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Pingfu Wang
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Zhang Liu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Qihang Yang
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Dongyu Li
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| |
Collapse
|
12
|
Wang Y, LeDue JM, Murphy TH. Multiscale imaging informs translational mouse modeling of neurological disease. Neuron 2022; 110:3688-3710. [PMID: 36198319 DOI: 10.1016/j.neuron.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Multiscale neurophysiology reveals that simple motor actions are associated with changes in neuronal firing in virtually every brain region studied. Accordingly, the assessment of focal pathology such as stroke or progressive neurodegenerative diseases must also extend widely across brain areas. To derive mechanistic information through imaging, multiple resolution scales and multimodal factors must be included, such as the structure and function of specific neurons and glial cells and the dynamics of specific neurotransmitters. Emerging multiscale methods in preclinical animal studies that span micro- to macroscale examinations fill this gap, allowing a circuit-based understanding of pathophysiological mechanisms. Combined with high-performance computation and open-source data repositories, these emerging multiscale and large field-of-view techniques include live functional ultrasound, multi- and single-photon wide-scale light microscopy, video-based miniscopes, and tissue-penetrating fiber photometry, as well as variants of post-mortem expansion microscopy. We present these technologies and outline use cases and data pipelines to uncover new knowledge within animal models of stroke, Alzheimer's disease, and movement disorders.
Collapse
Affiliation(s)
- Yundi Wang
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jeffrey M LeDue
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Timothy H Murphy
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
13
|
High-resolution micro-CT for 3D infarct characterization and segmentation in mice stroke models. Sci Rep 2022; 12:17471. [PMID: 36261475 PMCID: PMC9582034 DOI: 10.1038/s41598-022-21494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023] Open
Abstract
Characterization of brain infarct lesions in rodent models of stroke is crucial to assess stroke pathophysiology and therapy outcome. Until recently, the analysis of brain lesions was performed using two techniques: (1) histological methods, such as TTC (Triphenyltetrazolium chloride), a time-consuming and inaccurate process; or (2) MRI imaging, a faster, 3D imaging method, that comes at a high cost. In the last decade, high-resolution micro-CT for 3D sample analysis turned into a simple, fast, and cheaper solution. Here, we successfully describe the application of brain contrasting agents (Osmium tetroxide and inorganic iodine) for high-resolution micro-CT imaging for fine location and quantification of ischemic lesion and edema in mouse preclinical stroke models. We used the intraluminal transient MCAO (Middle Cerebral Artery Occlusion) mouse stroke model to identify and quantify ischemic lesion and edema, and segment core and penumbra regions at different time points after ischemia, by manual and automatic methods. In the transient-ischemic-attack (TIA) mouse model, we can quantify striatal myelinated fibers degeneration. Of note, whole brain 3D reconstructions allow brain atlas co-registration, to identify the affected brain areas, and correlate them with functional impairment. This methodology proves to be a breakthrough in the field, by providing a precise and detailed assessment of stroke outcomes in preclinical animal studies.
Collapse
|
14
|
Xue J, Singh S, Zhou Y, Perdomo-Pantoja A, Tian Y, Gupta N, Witham T, Grayson WL, Weihs TP. A biodegradable 3D woven magnesium-based scaffold for orthopedic implants. Biofabrication 2022; 14. [DOI: 10.1088/1758-5090/ac73b8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/26/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Porous Magnesium (Mg) is a promising biodegradable scaffold for treating critical-size bone defects, and as an essential element for human metabolism, Mg has shown sufficient biocompatibility. Its elastic moduli and yield strengths are closer to those of cortical bone than common, inert metallic implants, effectively reducing stress concentrations around host tissue as well as stress shielding. More importantly, Mg can degrade and be absorbed in the human body in a safe and controlled manner, thereby reducing the need for second surgeries to remove implants. The development of porous Mg scaffolds via conventional selective laser melting (SLM) techniques has been limited due to Mg’s low boiling point, high vapor pressures, high reactivity, and non-ideal microstructures in additively manufactured parts. Here we present an exciting alternative to conventional additive techniques: 3D weaving with Mg wires that have controlled chemistries and microstructures. The weaving process offers high throughput manufacturing as well as porous architectures that can be optimized for stiffness and porosity with topology optimization. Once woven, we dip-coat the weaves with polylactic acid (PLA) to enhance their strength and corrosion resistance. Following fabrication, we characterize their mechanical properties, corrosion behavior, and cell compatibility in vitro, and we use an intramuscular implantation model to evaluate their in vivo corrosion behavior and tissue response.
Collapse
|
15
|
Bonney SK, Coelho-Santos V, Huang SF, Takeno M, Kornfeld J, Keller A, Shih AY. Public Volume Electron Microscopy Data: An Essential Resource to Study the Brain Microvasculature. Front Cell Dev Biol 2022; 10:849469. [PMID: 35450291 PMCID: PMC9016339 DOI: 10.3389/fcell.2022.849469] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Electron microscopy is the primary approach to study ultrastructural features of the cerebrovasculature. However, 2D snapshots of a vascular bed capture only a small fraction of its complexity. Recent efforts to synaptically map neuronal circuitry using volume electron microscopy have also sampled the brain microvasculature in 3D. Here, we perform a meta-analysis of 7 data sets spanning different species and brain regions, including two data sets from the MICrONS consortium that have made efforts to segment vasculature in addition to all parenchymal cell types in mouse visual cortex. Exploration of these data have revealed rich information for detailed investigation of the cerebrovasculature. Neurovascular unit cell types (including, but not limited to, endothelial cells, mural cells, perivascular fibroblasts, microglia, and astrocytes) could be discerned across broad microvascular zones. Image contrast was sufficient to identify subcellular details, including endothelial junctions, caveolae, peg-and-socket interactions, mitochondria, Golgi cisternae, microvilli and other cellular protrusions of potential significance to vascular signaling. Additionally, non-cellular structures including the basement membrane and perivascular spaces were visible and could be traced between arterio-venous zones along the vascular wall. These explorations revealed structural features that may be important for vascular functions, such as blood-brain barrier integrity, blood flow control, brain clearance, and bioenergetics. They also identified limitations where accuracy and consistency of segmentation could be further honed by future efforts. The purpose of this article is to introduce these valuable community resources within the framework of cerebrovascular research. We do so by providing an assessment of their vascular contents, identifying features of significance for further study, and discussing next step ideas for refining vascular segmentation and analysis.
Collapse
Affiliation(s)
- Stephanie K Bonney
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States
| | - Vanessa Coelho-Santos
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Marc Takeno
- Allen Institute for Brain Science, Seattle, WA, United States
| | | | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Lu L, Liu X, Bian K, Sled JG, Monson K, Brown A, Mao H. The Effect of 3D Whole, Major, and Small Vasculature On Mouse Brain Strain Under Both Diffuse and Focal Brain Injury Loading. J Biomech Eng 2022; 144:1133338. [PMID: 35079765 DOI: 10.1115/1.4053664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Indexed: 11/08/2022]
Abstract
Blood vessels are much stiffer than brain parenchyma and their effects in finite element (FE) brain models need to be investigated. Despite the publication of some comprehensive three-dimensional (3D) brain vasculature models, no mechanical model exists for the mouse brain vasculature. Moreover, how the vasculature affects the mechanical behavior of brain tissue remains controversial. Therefore, we developed FE mouse brain models with detailed 3D vasculature to investigate the effect of the vasculature on brain strains under both diffuse (closed-head impact) and focal injury (controlled cortical impact (CCI)) loading, two commonly laboratory models of traumatic brain injury. The effect of the vasculature was examined by comparing maximum principal strain in mouse brain FE models with and without the vasculature. On average, modeling comprehensive vasculature under diffuse injury loading reduced average brain strain predictions by 32% with non-linear elastic properties. Nearly three-fourths of the 32% strain reduction was attributable to the effects of the major branches of the vasculature. Meanwhile, during focal open-skull CCI injury loading, the contribution of the vasculature was limited, producing a less than 5% reduction in all cases. Overall, the vasculature, especially the major branches, increased the load-bearing capacity of the brain FE model and thus reduced brain strain predictions.
Collapse
Affiliation(s)
- Lihong Lu
- Mechanical and Materials Engineering, Faculty of Engineering, Western University, London ON Canada
| | - Xingyu Liu
- Mechanical and Materials Engineering, Faculty of Engineering, Western University, London ON Canada
| | - Kewei Bian
- Mechanical and Materials Engineering, Faculty of Engineering, Western University, London ON Canada
| | - John G Sled
- Hospital for Sick Children, Toronto Canada; Medical Biophysics, University of Toronto, Canada
| | - Ken Monson
- Mechanical Engineering, The University of Utah, Salt Lake City UT USA; Biomedical Engineering, University of Utah, Salt Lake City UT USA
| | - Arthur Brown
- Robarts Research Institute, Western University, London ON Canada
| | - Haojie Mao
- Mechanical and Materials Engineering, Faculty of Engineering, Western University, London ON Canada; School of Biomedical Engineering, Western University, London ON Canada
| |
Collapse
|
17
|
Rosenblum JS, Cappadona AJ, Lookian PP, Chandrashekhar V, Bryant JP, Chandrashekhar V, Zhao DY, Knutsen RH, Donahue DR, McGavern DB, Kozel BA, Heiss JD, Pacak K, Zhuang Z. Non-invasive in situ Visualization of the Murine Cranial Vasculature. CELL REPORTS METHODS 2022; 2:100151. [PMID: 35373177 PMCID: PMC8967186 DOI: 10.1016/j.crmeth.2021.100151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/29/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
Understanding physiologic and pathologic central nervous system function depends on our ability to map the entire in situ cranial vasculature and neurovascular interfaces. To accomplish this, we developed a non-invasive workflow to visualize murine cranial vasculature via polymer casting of vessels, iterative sample processing and micro-computed tomography, and automatic deformable image registration, feature extraction, and visualization. This methodology is applicable to any tissue and allows rapid exploration of normal and altered pathologic states.
Collapse
Affiliation(s)
| | - Anthony J. Cappadona
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pashayar P. Lookian
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jean-Paul Bryant
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David Y. Zhao
- Department of Neurosurgery, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Russell H. Knutsen
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle R. Donahue
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth A. Kozel
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Redenski I, Guo S, Machour M, Szklanny A, Landau S, Egozi D, Gabet Y, Levenberg S. Microcomputed Tomography-Based Analysis of Neovascularization within Bioengineered Vascularized Tissues. ACS Biomater Sci Eng 2022; 8:232-241. [PMID: 34905338 DOI: 10.1021/acsbiomaterials.1c01401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the field of tissue engineering, evaluating newly formed vascular networks is considered a fundamental step in deciphering the processes underlying tissue development. Several common modalities exist to study vessel network formation and function. However, a proper methodology that allows through three-dimensional visualization of neovessels in a reproducible manner is required. Here, we describe in-depth exploration, visualization, and analysis of vessels within newly formed tissues by utilizing a contrast agent perfusion protocol and high-resolution microcomputed tomography. Bioengineered constructs consisting of porous, biocompatible, and biodegradable scaffolds are loaded with cocultures of adipose-derived microvascular endothelial cells (HAMECs) and dental pulp stem cells (DPSCs) and implanted in a rat femoral bundle model. After 14 days of in vivo maturation, we performed the optimized perfusion protocol to allow host penetrating vascular visualization and assessment within neotissues. Following high-resolution microCT scanning of DPSC:HAMEC explants, we performed the volumetric and spatial analysis of neovasculature. Eventually, the process was repeated with a previously published coculture system for prevascularization based on adipose-derived mesenchymal stromal cells (MSCs) and HAMECs. Overall, our approach allows a comprehensive understanding of vessel organization during engraftment and development of neotissues.
Collapse
Affiliation(s)
- Idan Redenski
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Shaowei Guo
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
- The First Affiliated Hospital, Shantou University Medical College, Shantou 515000, China
| | - Majd Machour
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Ariel Szklanny
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Shira Landau
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Dana Egozi
- Department of Plastic and Reconstructive Surgery, Kaplan Hospital, Rehovot and the Hebrew University, Jerusalem 9190401, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
19
|
Szklanny AA, Machour M, Redenski I, Chochola V, Goldfracht I, Kaplan B, Epshtein M, Simaan Yameen H, Merdler U, Feinberg A, Seliktar D, Korin N, Jaroš J, Levenberg S. 3D Bioprinting of Engineered Tissue Flaps with Hierarchical Vessel Networks (VesselNet) for Direct Host-To-Implant Perfusion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102661. [PMID: 34510579 PMCID: PMC11468543 DOI: 10.1002/adma.202102661] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Indexed: 05/09/2023]
Abstract
Engineering hierarchical vasculatures is critical for creating implantable functional thick tissues. Current approaches focus on fabricating mesoscale vessels for implantation or hierarchical microvascular in vitro models, but a combined approach is yet to be achieved to create engineered tissue flaps. Here, millimetric vessel-like scaffolds and 3D bioprinted vascularized tissues interconnect, creating fully engineered hierarchical vascular constructs for implantation. Endothelial and support cells spontaneously form microvascular networks in bioprinted tissues using a human collagen bioink. Sacrificial molds are used to create polymeric vessel-like scaffolds and endothelial cells seeded in their lumen form native-like endothelia. Assembling endothelialized scaffolds within vascularizing hydrogels incites the bioprinted vasculature and endothelium to cooperatively create vessels, enabling tissue perfusion through the scaffold lumen. Using a cuffing microsurgery approach, the engineered tissue is directly anastomosed with a rat femoral artery, promoting a rich host vasculature within the implanted tissue. After two weeks in vivo, contrast microcomputer tomography imaging and lectin perfusion of explanted engineered tissues verify the host ingrowth vasculature's functionality. Furthermore, the hierarchical vessel network (VesselNet) supports in vitro functionality of cardiomyocytes. Finally, the proposed approach is expanded to mimic complex structures with native-like millimetric vessels. This work presents a novel strategy aiming to create fully-engineered patient-specific thick tissue flaps.
Collapse
Affiliation(s)
- Ariel A. Szklanny
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Majd Machour
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Idan Redenski
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Václav Chochola
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrno625 00Czech Republic
| | - Idit Goldfracht
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Ben Kaplan
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Mark Epshtein
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Haneen Simaan Yameen
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Uri Merdler
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Adam Feinberg
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Dror Seliktar
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Netanel Korin
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Josef Jaroš
- Cell and Tissue RegenerationInternational Clinical Research CenterSt. Anne's University Hospital BrnoBrno65691Czech Republic
| | - Shulamit Levenberg
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| |
Collapse
|
20
|
Lan S, Yang B, Migneault F, Turgeon J, Bourgault M, Dieudé M, Cardinal H, Hickey MJ, Patey N, Hébert MJ. Caspase-3-dependent peritubular capillary dysfunction is pivotal for the transition from acute to chronic kidney disease after acute ischemia-reperfusion injury. Am J Physiol Renal Physiol 2021; 321:F335-F351. [PMID: 34338031 DOI: 10.1152/ajprenal.00690.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 07/21/2021] [Indexed: 11/22/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a major risk factor for chronic renal failure. Caspase-3, an effector responsible for apoptosis execution, is activated within the peritubular capillary (PTC) in the early stage of IRI-induced acute kidney injury (AKI). Recently, we showed that caspase-3-dependent microvascular rarefaction plays a key role in fibrosis development after mild renal IRI. Here, we further characterized the role of caspase-3 in microvascular dysfunction and progressive renal failure in both mild and severe AKI, by performing unilateral renal artery clamping for 30/60 min with contralateral nephrectomy in wild-type (C57BL/6) or caspase-3-/- mice. In both forms of AKI, caspase-3-/- mice showed better long-term outcomes despite worse initial tubular injury. After 3 wk, they showed reduced PTC injury, decreased PTC collagen deposition and α-smooth muscle actin expression, and lower tubular injury scores compared with wild-type animals. Caspase-3-/- mice with severe IRI also showed better preservation of long-term renal function. Intravital imaging and microcomputed tomography revealed preserved PTC permeability and better terminal capillary density in caspase-3-/- mice. Collectively, these results demonstrate the pivotal importance of caspase-3 in regulating long-term renal function after IRI and establish the predominant role of PTC dysfunction as a major contributor to progressive renal dysfunction.NEW & NOTEWORTHY Our findings demonstrate the pivotal importance of caspase-3 in regulating renal microvascular dysfunction, fibrogenesis, and long-term renal impairment after acute kidney injury induced by ischemia-reperfusion injury. Furthermore, this study establishes the predominant role of peritubular capillary integrity as a major contributor to progressive renal dysfunction after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Shanshan Lan
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Bing Yang
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Francis Migneault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Julie Turgeon
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Maude Bourgault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Natacha Patey
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- Department of Pathology, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
21
|
PEG-modified gadolinium nanoparticles as contrast agents for in vivo micro-CT. Sci Rep 2021; 11:16603. [PMID: 34400681 PMCID: PMC8367985 DOI: 10.1038/s41598-021-95716-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Vascular research is largely performed in rodents with the goal of developing treatments for human disease. Micro-computed tomography (micro-CT) provides non-destructive three-dimensional imaging that can be used to study the vasculature of rodents. However, to distinguish vasculature from other soft tissues, long-circulating contrast agents are required. In this study, we demonstrated that poly(ethylene glycol) (PEG)-coated gadolinium nanoparticles can be used as a vascular contrast agent in micro-CT. The coated particles could be lyophilized and then redispersed in an aqueous solution to achieve 100 mg/mL of gadolinium. After an intravenous injection of the contrast agent into mice, micro-CT scans showed blood pool contrast enhancements of at least 200 HU for 30 min. Imaging and quantitative analysis of gadolinium in tissues showed the presence of contrast agent in clearance organs including the liver and spleen and very low amounts in other organs. In vitro cell culture experiments, subcutaneous injections, and analysis of mouse body weight suggested that the agents exhibited low toxicity. Histological analysis of tissues 5 days after injection of the contrast agent showed cytotoxicity in the spleen, but no abnormalities were observed in the liver, lungs, kidneys, and bladder.
Collapse
|
22
|
Hartung G, Badr S, Mihelic S, Dunn A, Cheng X, Kura S, Boas DA, Kleinfeld D, Alaraj A, Linninger AA. Mathematical synthesis of the cortical circulation for the whole mouse brain-part II: Microcirculatory closure. Microcirculation 2021; 28:e12687. [PMID: 33615601 DOI: 10.1111/micc.12687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/23/2020] [Accepted: 02/10/2021] [Indexed: 11/29/2022]
Abstract
Recent advancements in multiphoton imaging and vascular reconstruction algorithms have increased the amount of data on cerebrovascular circulation for statistical analysis and hemodynamic simulations. Experimental observations offer fundamental insights into capillary network topology but mainly within a narrow field of view typically spanning a small fraction of the cortical surface (less than 2%). In contrast, larger-resolution imaging modalities, such as computed tomography (CT) or magnetic resonance imaging (MRI), have whole-brain coverage but capture only larger blood vessels, overlooking the microscopic capillary bed. To integrate data acquired at multiple length scales with different neuroimaging modalities and to reconcile brain-wide macroscale information with microscale multiphoton data, we developed a method for synthesizing hemodynamically equivalent vascular networks for the entire cerebral circulation. This computational approach is intended to aid in the quantification of patterns of cerebral blood flow and metabolism for the entire brain. In part I, we described the mathematical framework for image-guided generation of synthetic vascular networks covering the large cerebral arteries from the circle of Willis through the pial surface network leading back to the venous sinuses. Here in part II, we introduce novel procedures for creating microcirculatory closure that mimics a realistic capillary bed. We demonstrate our capability to synthesize synthetic vascular networks whose morphometrics match empirical network graphs from three independent state-of-the-art imaging laboratories using different image acquisition and reconstruction protocols. We also successfully synthesized twelve vascular networks of a complete mouse brain hemisphere suitable for performing whole-brain blood flow simulations. Synthetic arterial and venous networks with microvascular closure allow whole-brain hemodynamic predictions. Simulations across all length scales will potentially illuminate organ-wide supply and metabolic functions that are inaccessible to models reconstructed from image data with limited spatial coverage.
Collapse
Affiliation(s)
- Grant Hartung
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Shoale Badr
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Samuel Mihelic
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Xiaojun Cheng
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - David Kleinfeld
- Department of Physics, University of California San Diego, San Diego, California, USA
| | - Ali Alaraj
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Andreas A Linninger
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Neurosurgery, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
23
|
Leyssens L, Pestiaux C, Kerckhofs G. A Review of Ex Vivo X-ray Microfocus Computed Tomography-Based Characterization of the Cardiovascular System. Int J Mol Sci 2021; 22:3263. [PMID: 33806852 PMCID: PMC8004599 DOI: 10.3390/ijms22063263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular malformations and diseases are common but complex and often not yet fully understood. To better understand the effects of structural and microstructural changes of the heart and the vasculature on their proper functioning, a detailed characterization of the microstructure is crucial. In vivo imaging approaches are noninvasive and allow visualizing the heart and the vasculature in 3D. However, their spatial image resolution is often too limited for microstructural analyses, and hence, ex vivo imaging is preferred for this purpose. Ex vivo X-ray microfocus computed tomography (microCT) is a rapidly emerging high-resolution 3D structural imaging technique often used for the assessment of calcified tissues. Contrast-enhanced microCT (CE-CT) or phase-contrast microCT (PC-CT) improve this technique by additionally allowing the distinction of different low X-ray-absorbing soft tissues. In this review, we present the strengths of ex vivo microCT, CE-CT and PC-CT for quantitative 3D imaging of the structure and/or microstructure of the heart, the vasculature and their substructures in healthy and diseased state. We also discuss their current limitations, mainly with regard to the contrasting methods and the tissue preparation.
Collapse
Affiliation(s)
- Lisa Leyssens
- Institute of Mechanics, Materials, and Civil Engineering, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; (L.L.); (C.P.)
- Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Woluwe-Saint-Lambert, Belgium
| | - Camille Pestiaux
- Institute of Mechanics, Materials, and Civil Engineering, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; (L.L.); (C.P.)
- Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Woluwe-Saint-Lambert, Belgium
| | - Greet Kerckhofs
- Institute of Mechanics, Materials, and Civil Engineering, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; (L.L.); (C.P.)
- Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Woluwe-Saint-Lambert, Belgium
- Department of Materials Engineering, Katholieke Universiteit Leuven, 3001 Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| |
Collapse
|
24
|
Contrast enhanced X-ray computed tomography imaging of amyloid plaques in Alzheimer disease rat model on lab based micro CT system. Sci Rep 2021; 11:5999. [PMID: 33727592 PMCID: PMC7966753 DOI: 10.1038/s41598-021-84579-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
Amyloid plaques are small (~ 50 μm), highly-dense aggregates of amyloid beta (Aβ) protein in brain tissue, supposed to play a key role in pathogenesis of Alzheimer’s disease (AD). Plaques´ in vivo detection, spatial distribution and quantitative characterization could be an essential marker in diagnostics and evaluation of AD progress. However, current imaging methods in clinics possess substantial limits in sensitivity towards Aβ plaques to play a considerable role in AD screening. Contrast enhanced X-ray micro computed tomography (micro CT) is an emerging highly sensitive imaging technique capable of high resolution visualization of rodent brain. In this study we show the absorption based contrast enhanced X-ray micro CT imaging is viable method for detection and 3D analysis of Aβ plaques in transgenic rodent models of Alzheimer’s disease. Using iodine contrasted brain tissue isolated from the Tg-F344-AD rat model we show the micro CT imaging is capable of precise imaging of Aβ plaques, making possible to further analyze various aspects of their 3D spatial distribution and other properties.
Collapse
|
25
|
Abstract
The quantitative analysis of blood vessel networks is an important component in many animal models of disease. We describe a nondestructive technique for blood vessel imaging that visualizes in situ vasculature in harvested tissues. The method allows for further analysis of the same tissues with histology and other methods that can be performed on fixed tissue. Consequently, it can easily be incorporated upstream to analysis methods to augment these with a three-dimensional reconstruction of the vascular network in the tissues to be analyzed. The method combines iodine-enhanced micro-computed tomography with a deep learning algorithm to segment vasculature within tissues. The procedure is relatively simple and can provide insight into complex changes in the vascular structure in the tissues.
Collapse
|
26
|
Hill LK, Hoang DM, Chiriboga LA, Wisniewski T, Sadowski MJ, Wadghiri YZ. Detection of Cerebrovascular Loss in the Normal Aging C57BL/6 Mouse Brain Using in vivo Contrast-Enhanced Magnetic Resonance Angiography. Front Aging Neurosci 2020; 12:585218. [PMID: 33192479 PMCID: PMC7606987 DOI: 10.3389/fnagi.2020.585218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/28/2022] Open
Abstract
Microvascular rarefaction, or the decrease in vascular density, has been described in the cerebrovasculature of aging humans, rats, and, more recently, mice in the presence and absence of age-dependent diseases. Given the wide use of mice in modeling age-dependent human diseases of the cerebrovasculature, visualization, and quantification of the global murine cerebrovasculature is necessary for establishing the baseline changes that occur with aging. To provide in vivo whole-brain imaging of the cerebrovasculature in aging C57BL/6 mice longitudinally, contrast-enhanced magnetic resonance angiography (CE-MRA) was employed using a house-made gadolinium-bearing micellar blood pool agent. Enhancement in the vascular space permitted quantification of the detectable, or apparent, cerebral blood volume (aCBV), which was analyzed over 2 years of aging and compared to histological analysis of the cerebrovascular density. A significant loss in the aCBV was detected by CE-MRA over the aging period. Histological analysis via vessel-probing immunohistochemistry confirmed a significant loss in the cerebrovascular density over the same 2-year aging period, validating the CE-MRA findings. While these techniques use widely different methods of assessment and spatial resolutions, their comparable findings in detected vascular loss corroborate the growing body of literature describing vascular rarefaction aging. These findings suggest that such age-dependent changes can contribute to cerebrovascular and neurodegenerative diseases, which are modeled using wild-type and transgenic laboratory rodents.
Collapse
Affiliation(s)
- Lindsay K. Hill
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, United States
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Dung Minh Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| | - Luis A. Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
| | - Martin J. Sadowski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| | - Youssef Z. Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
27
|
Advances in imaging feto-placental vasculature: new tools to elucidate the early life origins of health and disease. J Dev Orig Health Dis 2020; 12:168-178. [PMID: 32746961 DOI: 10.1017/s2040174420000720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Optimal placental function is critical for fetal development, and therefore a crucial consideration for understanding the developmental origins of health and disease (DOHaD). The structure of the fetal side of the placental vasculature is an important determinant of fetal growth and cardiovascular development. There are several imaging modalities for assessing feto-placental structure including stereology, electron microscopy, confocal microscopy, micro-computed tomography, light-sheet microscopy, ultrasonography and magnetic resonance imaging. In this review, we present current methodologies for imaging feto-placental vasculature morphology ex vivo and in vivo in human and experimental models, their advantages and limitations and how these provide insight into placental function and fetal outcomes. These imaging approaches add important perspective to our understanding of placental biology and have potential to be new tools to elucidate a deeper understanding of DOHaD.
Collapse
|
28
|
Kuo W, Le NA, Spingler B, Wenger RH, Kipar A, Hetzel U, Schulz G, Müller B, Kurtcuoglu V. Simultaneous Three-Dimensional Vascular and Tubular Imaging of Whole Mouse Kidneys With X-ray μCT. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:731-740. [PMID: 32627730 DOI: 10.1017/s1431927620001725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Concurrent three-dimensional imaging of the renal vascular and tubular systems on the whole-kidney scale with capillary level resolution is labor-intensive and technically difficult. Approaches based on vascular corrosion casting and X-ray micro computed tomography (μCT), for example, suffer from vascular filling artifacts and necessitate imaging with an additional modality to acquire tubules. In this work, we report on a new sample preparation, image acquisition, and quantification protocol for simultaneous vascular and tubular μCT imaging of whole, uncorroded mouse kidneys. The protocol consists of vascular perfusion with the water-soluble, aldehyde-fixable, polymeric X-ray contrast agent XlinCA, followed by laboratory-source μCT imaging and structural analysis using the freely available Fiji/ImageJ software. We achieved consistent filling of the entire capillary bed and staining of the tubules in the cortex and outer medulla. After imaging at isotropic voxel sizes of 3.3 and 4.4 μm, we segmented vascular and tubular systems and quantified luminal volumes, surface areas, diffusion distances, and vessel path lengths. This protocol permits the analysis of vascular and tubular parameters with higher reliability than vascular corrosion casting, less labor than serial sectioning and leaves tissue intact for subsequent histological examination with light and electron microscopy.
Collapse
Affiliation(s)
- Willy Kuo
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Ngoc An Le
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Bernhard Spingler
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Roland H Wenger
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Anja Kipar
- University of Zurich, Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Udo Hetzel
- University of Zurich, Electron Microscopy Unit, Institute of Veterinary Pathology, Vetsuisse Faculty, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Georg Schulz
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Bert Müller
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Vartan Kurtcuoglu
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, Zurich Center for Integrative Human Physiology, 8057Zurich, Switzerland
| |
Collapse
|
29
|
Bernstein DL, Rom S. Let-7g* and miR-98 Reduce Stroke-Induced Production of Proinflammatory Cytokines in Mouse Brain. Front Cell Dev Biol 2020; 8:632. [PMID: 32766248 PMCID: PMC7379105 DOI: 10.3389/fcell.2020.00632] [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: 05/06/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is a debilitating illness facing healthcare today, affecting over 800,000 people and causing over 140,000 deaths each year in the United States. Despite being the third-leading cause of death, very few treatments currently exist for stroke. Often, during an ischemic attack, the blood-brain barrier (BBB) is significantly damaged, which can lead to altered interactions with the immune system, and greatly worsen the damage from a stroke. The impaired, BBB promotes the infiltration of peripheral inflammatory cells into the brain, secreting deleterious mediators (cytokines/chemokines) and resulting in permanent barrier injury. let-7 microRNAs (miRs) are critical for regulating immune responses within the BBB, particularly after ischemic stroke. We have previously shown how transient stroke decreases expression of multiple let-7 miRs, and that restoration of expression confers significant neuroprotection, reduction in brain infiltration by neutrophils, monocytes and T cells. However, the specific mechanisms of action of let-7 miRs remain unexplored, though emerging evidence implicates a range of impacts on cytokines. In the current study, we evaluate the impacts of miR-98 and let-7g* on targeting of cytokine mRNAs, cytokine release following ischemic stroke, and cell-specific changes to the neurovascular space. We determined that miR-98 specifically targets IP-10, while let-7g* specifically aims IL-8, and attenuates their levels. Both produce strong impacts on CCL2 and CCL5. Further, let-7g* strongly improves neurovascular perfusion following ischemic stroke. Together, the results of the study indicate that let-7 miRs are critical for mediating endothelial-immune reactions and improving recovery following ischemic stroke.
Collapse
Affiliation(s)
- David L Bernstein
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
30
|
Structural Variant in Mitochondrial-Associated Gene (MRPL3) Induces Adult-Onset Neurodegeneration with Memory Impairment in the Mouse. J Neurosci 2020; 40:4576-4585. [PMID: 32341096 DOI: 10.1523/jneurosci.0013-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/31/2022] Open
Abstract
An impediment to the development of effective therapies for neurodegenerative disease is that available animal models do not reproduce important clinical features such as adult-onset and stereotypical patterns of progression. Using in vivo magnetic resonance imaging and behavioral testing to study male and female decrepit mice, we found a stereotypical neuroanatomical pattern of progression of the lesion along the limbic system network and an associated memory impairment. Using structural variant analysis, we identified an intronic mutation in a mitochondrial-associated gene (Mrpl3) that is responsible for the decrepit phenotype. While the function of this gene is unknown, embryonic lethality in Mrpl3 knock-out mice suggests it is critical for early development. The observation that a mutation linked to energy metabolism precipitates a pattern of neurodegeneration via cell death across disparate but linked brain regions may explain how stereotyped patterns of neurodegeneration arise in humans or define a not yet identified human disease.SIGNIFICANCE STATEMENT The development of novel therapies for adult-onset neurodegenerative disease has been impeded by the limitations of available animal models in reproducing many of the clinical features. Here, we present a novel spontaneous mutation in a mitochondrial-associated gene in a mouse (termed decrepit) that results in adult-onset neurodegeneration with a stereotypical neuroanatomical pattern of progression and an associated memory impairment. The decrepit mouse model may represent a heretofore undiagnosed human disease and could serve as a new animal model to study neurodegenerative disease.
Collapse
|
31
|
Le NA, Kuo W, Müller B, Kurtcuoglu V, Spingler B. Crosslinkable polymeric contrast agent for high-resolution X-ray imaging of the vascular system. Chem Commun (Camb) 2020; 56:5885-5888. [DOI: 10.1039/c9cc09883f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A contrast agent for X-ray micro computed tomography (μCT), called XlinCA, that combines reliable perfusion and permanent retention and contrast properties, was developed for ex vivo imaging.
Collapse
Affiliation(s)
- Ngoc An Le
- Department of Chemistry
- University of Zurich
- 8057 Zurich
- Switzerland
| | - Willy Kuo
- Institute of Physiology
- University of Zurich
- 8057 Zurich
- Switzerland
- National Centre of Competence in Research
| | - Bert Müller
- Biomaterials Science Center
- Department of Biomedical Engineering
- University of Basel
- 4123 Allschwil
- Switzerland
| | - Vartan Kurtcuoglu
- Institute of Physiology
- University of Zurich
- 8057 Zurich
- Switzerland
- National Centre of Competence in Research
| | | |
Collapse
|
32
|
Cahill LS, Pilmeyer J, Yu LX, Steinman J, Hare GMT, Kassner A, Macgowan CK, Sled JG. Ultrasound Detection of Abnormal Cerebrovascular Morphology in a Mouse Model of Sickle Cell Disease Based on Wave Reflection. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3269-3278. [PMID: 31563480 DOI: 10.1016/j.ultrasmedbio.2019.08.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/09/2019] [Accepted: 08/23/2019] [Indexed: 06/10/2023]
Abstract
Sickle cell disease (SCD) is associated with a high risk of stroke, and affected individuals often have focal brain lesions termed silent cerebral infarcts. The mechanisms leading to these types of injuries are at present poorly understood. Our group has recently demonstrated a non-invasive measurement of cerebrovascular impedance and wave reflection in mice using high-frequency ultrasound in the common carotid artery. To better understand the pathophysiology in SCD, we used this approach in combination with micro-computed tomography to investigate changes in cerebrovascular morphology in the Townes mouse model of SCD. Relative to controls, the SCD mice demonstrated the following: (i) increased carotid artery diameter, blood flow and vessel wall thickness; (ii) elevated pulse wave velocity; (iii) increased reflection coefficient; and (iv) an increase in the total number of vessel segments in the brain. This study highlights the potential for wave reflection to aid the non-invasive clinical assessment of vascular pathology in SCD.
Collapse
Affiliation(s)
- Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Jesper Pilmeyer
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa X Yu
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joe Steinman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gregory M T Hare
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Kassner
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Christopher K Macgowan
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Saffari TM, Mathot F, Bishop AT, Shin AY. New methods for objective angiogenesis evaluation of rat nerves using microcomputed tomography scanning and conventional photography. Microsurgery 2019; 40:370-376. [PMID: 31758730 DOI: 10.1002/micr.30537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/01/2019] [Accepted: 11/08/2019] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Nerve regeneration involves multiple processes, which enhance blood supply that can be promoted by growth factors. Currently, tools are lacking to visualize the vascularization patterns in transplanted nerves in vivo. The purpose of this study was to describe three-dimensional visualization of the vascular system in the rat sciatic nerve and to quantify angiogenesis of nerve reconstruction. MATERIALS AND METHODS In 12 Lewis rats (weighing 250-300 g), 10 mm sciatic nerve gaps were repaired with ipsilateral reversed autologous nerve grafts. At 12 and 16 weeks of sacrifice, Microfil® contrast compound was injected in the aorta. Nerve autografts (N = 12) and contralateral untreated nerves (N = 12) were harvested and cleared while preserving the vasculature. The amount of vascularization was measured by quantifying the vascular surface area using conventional photography (two-dimensional) and the vascular volume was calculated with microcomputed tomography (three-dimensional). For each measurement, a vessel/nerve area ratio was calculated and expressed in percentages (vessel%). RESULTS The vascular volume measured 3.53 ± 0.43% in autografts and 4.83 ± 0.45% vessels in controls at 12 weeks and 4.95 ± 0.44% and 6.19 ± 0.29% vessels at 16 weeks, respectively. The vascular surface area measured 25.04 ± 2.77% in autografts and 26.87 ± 2.13% vessels in controls at 12 weeks, and 28.11 ± 3.47% and 33.71 ± 2.60% vessels at 16 weeks, respectively. The correlation between both methods was statistically significant (p = .049). CONCLUSIONS Both methods are considered to successfully reflect the degree of vascularization. Application of this technique could be used to visualize and objectively quantify angiogenesis of the transplanted nerve graft. Moreover, this simple method is easily reproducible and could be extrapolated to any other desired target organ ex vivo in small animals to investigate the vascular network.
Collapse
Affiliation(s)
- Tiam M Saffari
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Plastic-, Reconstructive- and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Femke Mathot
- Department of Plastic Surgery, Radboud University, Nijmegen, The Netherlands
| | - Allen T Bishop
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Alexander Y Shin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
34
|
Quintana DD, Lewis SE, Anantula Y, Garcia JA, Sarkar SN, Cavendish JZ, Brown CM, Simpkins JW. The cerebral angiome: High resolution MicroCT imaging of the whole brain cerebrovasculature in female and male mice. Neuroimage 2019; 202:116109. [PMID: 31446129 PMCID: PMC6942880 DOI: 10.1016/j.neuroimage.2019.116109] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 01/09/2023] Open
Abstract
The cerebrovascular system provides crucial functions that maintain metabolic and homeostatic states of the brain. Despite its integral role of supporting cerebral viability, the topological organization of these networks remains largely uncharacterized. This void in our knowledge surmises entirely from current technological limitations that prevent the capturing of data through the entire depth of the brain. We report high-resolution reconstruction and analysis of the complete vascular network of the entire brain at the capillary level in adult female and male mice using a vascular corrosion cast procedure. Vascular network analysis of the whole brain revealed sex-related differences of vessel hierarchy. In addition, region-specific network analysis demonstrated different patterns of angioarchitecture between brain subregions and sex. Furthermore, our group is the first to provide a three-dimensional analysis of the angioarchitecture and network organization in a single reconstructed tomographic data set that encompasses all hierarchy of vessels in the brain of the adult mouse.
Collapse
Affiliation(s)
- D D Quintana
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S E Lewis
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - Y Anantula
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J A Garcia
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S N Sarkar
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J Z Cavendish
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - C M Brown
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J W Simpkins
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
35
|
Nyberg E, Farris A, O'Sullivan A, Rodriguez R, Grayson W. Comparison of Stromal Vascular Fraction and Passaged Adipose-Derived Stromal/Stem Cells as Point-of-Care Agents for Bone Regeneration. Tissue Eng Part A 2019; 25:1459-1469. [DOI: 10.1089/ten.tea.2018.0341] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Ethan Nyberg
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ashley Farris
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aine O'Sullivan
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
36
|
Deng Y, Rowe KJ, Chaudhary KR, Yang A, Mei SHJ, Stewart DJ. Optimizing imaging of the rat pulmonary microvasculature by micro-computed tomography. Pulm Circ 2019; 9:2045894019883613. [PMID: 31700608 PMCID: PMC6823983 DOI: 10.1177/2045894019883613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022] Open
Abstract
Micro-computed tomography (micro-CT) is used in pre-clinical research to generate high-resolution three-dimensional (3D) images of organs and tissues. When combined with intravascular contrast agents, micro-CT can provide 3D visualization and quantification of vascular networks in many different organs. However, the lungs present a particular challenge for contrast perfusion due to the complexity and fragile nature of the lung microcirculation. The protocol described here has been optimized to achieve consistent lung perfusion of the microvasculature to vessels < 20 microns in both normal and pulmonary arterial hypertension rats. High-resolution 3D micro-CT imaging can be used to better visualize changes in 3D architecture of the lung microcirculation in pulmonary vascular disease and to assess the impact of therapeutic strategies on microvascular structure in animal models of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Yupu Deng
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Katelynn J Rowe
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Ketul R Chaudhary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Anli Yang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
37
|
Hong SH, Herman AM, Stephenson JM, Wu T, Bahadur AN, Burns AR, Marrelli SP, Wythe JD. Development of barium-based low viscosity contrast agents for micro CT vascular casting: Application to 3D visualization of the adult mouse cerebrovasculature. J Neurosci Res 2019; 98:312-324. [PMID: 31630455 DOI: 10.1002/jnr.24539] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/09/2019] [Accepted: 09/23/2019] [Indexed: 11/07/2022]
Abstract
Recent advances in three-dimensional (3D) fluorescence microscopy offer the ability to image the entire vascular network in entire organs, or even whole animals. However, these imaging modalities rely on either endogenous fluorescent reporters or involved immunohistochemistry protocols, as well as optical clearing of the tissue and refractive index matching. Conversely, X-ray-based 3D imaging modalities, such as micro CT, can image non-transparent samples, at high resolution, without requiring complicated or expensive immunolabeling and clearing protocols, or fluorescent reporters. Here, we compared two "homemade" barium-based contrast agents to the field standard, lead-containing Microfil, for micro-computed tomography (micro CT) imaging of the adult mouse cerebrovasculature. The perfusion pressure required for uniform vessel filling was significantly lower with the barium-based contrast agents compared to the polymer-based Microfil. Accordingly, the barium agents showed no evidence of vascular distension or rupture, common problems associated with Microfil. Compellingly, perfusion of an aqueous BaCl2 /gelatin mixture yielded equal or superior visualization of the cerebrovasculature by micro CT compared to Microfil. However, phosphate-containing buffers and fixatives were incompatible with BaCl2 due to the formation of unwanted precipitates. X-ray attenuation of the vessels also decreased overtime, as the BaCl2 appeared to gradually diffuse into surrounding tissues. A second, unique formulation composed of BaSO4 microparticles, generated in-house by mixing BaCl2 and MgSO4 , suffered none of these drawbacks. These microparticles, however, were unable to pass small diameter capillary vessels, conveniently labeling only the arterial cerebrovasculature. In summary, we present an affordable, robust, low pressure, non-toxic, and straightforward methodology for 3D visualization of the cerebrovasculature.
Collapse
Affiliation(s)
- Sung-Ha Hong
- Department of Neurology, McGovern Medical School at UT Health, Houston, TX, USA
| | - Alexander M Herman
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | - Ting Wu
- Department of Neurology, McGovern Medical School at UT Health, Houston, TX, USA
| | | | - Alan R Burns
- College of Optometry, University of Houston, Houston, TX, USA
| | - Sean P Marrelli
- Department of Neurology, McGovern Medical School at UT Health, Houston, TX, USA
| | - Joshua D Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
38
|
Mrzílková J, Patzelt M, Gallina P, Wurst Z, Šeremeta M, Dudák J, Krejčí F, Žemlička J, Musil V, Karch J, Rosina J, Zach P. Imaging of Mouse Brain Fixated in Ethanol in Micro-CT. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2054262. [PMID: 31392208 PMCID: PMC6662504 DOI: 10.1155/2019/2054262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/07/2019] [Accepted: 06/20/2019] [Indexed: 12/25/2022]
Abstract
Micro-CT imaging is a well-established morphological method for the visualization of animal models. We used ethanol fixation of the mouse brains to perform high-resolution micro-CT scans showing in great details brain grey and white matters. It was possible to identify more than 50 neuroanatomical structures on the 5 selected coronal sections. Among white matter structures, we identified fornix, medial lemniscus, crossed tectospinal pathway, mammillothalamic tract, and the sensory root of the trigeminal ganglion. Among grey matter structures, we identified basal nuclei, habenular complex, thalamic nuclei, amygdala, subparts of hippocampal formation, superior colliculi, Edinger-Westphal nucleus, and others. We suggest that micro-CT of the mouse brain could be used for neurohistological lesions evaluation as an alternative to classical neurohistology because it does not destroy brain tissue.
Collapse
Affiliation(s)
- Jana Mrzílková
- Specialized Laboratory of Experimental Imaging Third Faculty of Medicine, Charles University, Institute of Experimental and Applied Physics and Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Matěj Patzelt
- Specialized Laboratory of Experimental Imaging Third Faculty of Medicine, Charles University, Institute of Experimental and Applied Physics and Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pasquale Gallina
- Department of Surgery and Translational Medicine, Neurosurgery Unit, Florence School of Neurosurgery, University of Florence, Florence, Italy
| | - Zdeněk Wurst
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Šeremeta
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Dudák
- Institute of Experimental and Applied Physics, Czech Technical University, Prague, Czech Republic
- Czech Technical University in Prague, Faculty of Biomedical Engineering, Kladno, Czech Republic
| | - František Krejčí
- Institute of Experimental and Applied Physics, Czech Technical University, Prague, Czech Republic
| | - Jan Žemlička
- Institute of Experimental and Applied Physics, Czech Technical University, Prague, Czech Republic
| | - Vladimír Musil
- Specialized Laboratory of Experimental Imaging Third Faculty of Medicine, Charles University, Institute of Experimental and Applied Physics and Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Centre of Scientific Information, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jakub Karch
- Institute of Experimental and Applied Physics, Czech Technical University, Prague, Czech Republic
| | - Jozef Rosina
- Czech Technical University in Prague, Faculty of Biomedical Engineering, Kladno, Czech Republic
- Department of Medical Biophysics and Informatics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Zach
- Specialized Laboratory of Experimental Imaging Third Faculty of Medicine, Charles University, Institute of Experimental and Applied Physics and Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
39
|
Li T, Liu CJ, Akkin T. Contrast-enhanced serial optical coherence scanner with deep learning network reveals vasculature and white matter organization of mouse brain. NEUROPHOTONICS 2019; 6:035004. [PMID: 31338386 PMCID: PMC6646884 DOI: 10.1117/1.nph.6.3.035004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 07/02/2019] [Indexed: 06/01/2023]
Abstract
Optical coherence tomography provides volumetric reconstruction of brain structure with micrometer resolution. Gray matter and white matter can be highlighted using conventional and polarization-based contrasts; however, vasculature in ex-vivo fixed brain has not been investigated at large scale due to lack of intrinsic contrast. We present contrast enhancement to visualize the vasculature by perfusing titanium dioxide particles transcardially into the mouse vascular system. The brain, after dissection and fixation, is imaged by a serial optical coherence scanner. Accumulation of particles in blood vessels generates distinguishable optical signals. Among these, the cross-polarization images reveal the vasculature organization remarkably well. The conventional and polarization-based contrasts are still available for probing the gray matter and white matter structures. The segmentation and reconstruction of the vasculature are presented by using a deep learning algorithm. Axonal fiber pathways in the mouse brain are delineated by utilizing the retardance and optic axis orientation contrasts. This is a low-cost method that can be further developed to study neurovascular diseases and brain injury in animal models.
Collapse
Affiliation(s)
- Tianqi Li
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Chao J. Liu
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Taner Akkin
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| |
Collapse
|
40
|
Adult Pgf -/- mice behaviour and neuroanatomy are altered by neonatal treatment with recombinant placental growth factor. Sci Rep 2019; 9:9285. [PMID: 31243296 PMCID: PMC6594955 DOI: 10.1038/s41598-019-45824-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Offspring of preeclamptic pregnancies have cognitive alterations. Placental growth factor (PGF), is low in preeclampsia; reduced levels may affect brain development. PGF-null mice differ from normal congenic controls in cerebrovasculature, neuroanatomy and behavior. Using brain imaging and behavioral testing, we asked whether developmentally asynchronous (i.e. neonatal) PGF supplementation alters the vascular, neuroanatomic and/or behavioral status of Pgf−/− mice at adulthood. C57BL/6-Pgf−/− pups were treated intraperitoneally on postnatal days 1–10 with vehicle or PGF at 10 pg/g, 70 pg/g or 700 pg/g. These mice underwent behavioral testing and perfusion for MRI and analysis of retinal vasculature. A second cohort of vehicle- or PGF-treated mice was perfused for micro-CT imaging. 10 pg/g PGF-treated mice exhibited less locomotor activity and greater anxiety-like behavior relative to vehicle-treated mice. Depressive-like behavior showed a sex-specific, dose-dependent decrease and was lowest in 700 pg/g PGF-treated females relative to vehicle-treated females. Spatial learning did not differ. MRI revealed smaller volume of three structures in the 10 pg/g group, larger volume of seven structures in the 70 pg/g group and smaller volume of one structure in the 700 pg/g group. No cerebral or retinal vascular differences were detected. Overall, neonatal PGF replacement altered behavior and neuroanatomy of adult Pgf−/− mice.
Collapse
|
41
|
Peeters TH, Kobus T, Breukels V, Lenting K, Veltien A, Heerschap A, Scheenen TWJ. Imaging Hyperpolarized Pyruvate and Lactate after Blood-Brain Barrier Disruption with Focused Ultrasound. ACS Chem Neurosci 2019; 10:2591-2601. [PMID: 30873831 PMCID: PMC6523999 DOI: 10.1021/acschemneuro.9b00085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
![]()
Imaging of hyperpolarized 13C-labeled substrates has
emerged as an important magnetic resonance (MR) technique to study
metabolic pathways in real time in vivo. Even though
this technique has found its way to clinical trials, in vivo dynamic nuclear polarization is still mostly applied in preclinical
models. Its tremendous increase in signal-to-noise ratio (SNR) overcomes
the intrinsically low MR sensitivity of the 13C nucleus
and allows real-time metabolic imaging in small structures like the
mouse brain. However, applications in brain research are limited as
delivery of hyperpolarized compounds is restrained by the blood–brain
barrier (BBB). A local noninvasive disruption of the BBB could facilitate
delivery of hyperpolarized substrates and create opportunities to
study metabolic pathways in the brain that are generally not within
reach. In this work, we designed a setup to apply BBB disruption in
the mouse brain by MR-guided focused ultrasound (FUS) prior to MR
imaging of 13C-enriched hyperpolarized [1-13C]-pyruvate and its conversion to [1-13C]-lactate. To
overcome partial volume issues, we optimized a fast multigradient-echo
imaging method (temporal resolution of 2.4 s) with an in-plane spatial
resolution of 1.6 × 1.6 mm2, without the need of processing
large amounts of spectroscopic data. We demonstrated the feasibility
to apply 13C imaging in less than 1 h after FUS treatment
and showed a locally disrupted BBB during the time window of the whole
experiment. From detected hyperpolarized pyruvate and lactate signals
in both FUS-treated and untreated mice, we conclude that even at high
spatial resolution, signals from the blood compartment dominate in
the 13C images, leaving the interpretation of hyperpolarized
signals in the mouse brain challenging.
Collapse
Affiliation(s)
- Tom H. Peeters
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Thiele Kobus
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Vincent Breukels
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Krissie Lenting
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Andor Veltien
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Tom W. J. Scheenen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
42
|
Linninger A, Hartung G, Badr S, Morley R. Mathematical synthesis of the cortical circulation for the whole mouse brain-part I. theory and image integration. Comput Biol Med 2019; 110:265-275. [PMID: 31247510 DOI: 10.1016/j.compbiomed.2019.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/25/2019] [Accepted: 05/04/2019] [Indexed: 12/19/2022]
Abstract
Microcirculation plays a significant role in cerebral metabolism and blood flow control, yet explaining and predicting functional mechanisms remains elusive because it is difficult to make physiologically accurate mathematical models of the vascular network. As a precursor to the human brain, this paper presents a computational framework for synthesizing anatomically accurate network models for the cortical blood supply in mouse. It addresses two critical deficiencies in cerebrovascular modeling. At the microscopic length scale of individual capillaries, we present a novel synthesis method for building anatomically consistent capillary networks with loops and anastomoses (=microcirculatory closure). This overcomes shortcomings in existing algorithms which are unable to create closed circulatory networks. A second critical innovation allows the incorporation of detailed anatomical features from image data into vascular growth. Specifically, computed tomography and two photon laser scanning microscopy data are input into the novel synthesis algorithm to build the cortical circulation for the entire mouse brain in silico. Computer predictions of blood flow and oxygen exchange executed on synthetic large-scale network models are expected to elucidate poorly understood functional mechanisms of the cerebral circulation.
Collapse
Affiliation(s)
- Andreas Linninger
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA; Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL, USA.
| | - Grant Hartung
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Shoale Badr
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Ryan Morley
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
43
|
Angiogenesis involvement by octacalcium phosphate-gelatin composite-driven bone regeneration in rat calvaria critical-sized defect. Acta Biomater 2019; 88:514-526. [PMID: 30776505 DOI: 10.1016/j.actbio.2019.02.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/26/2019] [Accepted: 02/14/2019] [Indexed: 12/29/2022]
Abstract
Effect of octacalcium phosphate/gelatin composite (OCP/Gel) on angiogenesis was studied by its implantation in rat calvaria critical-sized defect in relation to bone regeneration for 2 and 4 weeks. The implantation of OCP/Gel disks was analyzed by radiomorphometry using a radiopaque material perfusion (Microfil®) method and histomorphometry by hematoxylin and eosin-staining before and after the decalcification. Effect of the OCP dose in the range up to 4 mg per well on the capillary-like tube formation by human umbilical vein endothelial cells (HUVECs) was also examined in a transwell cell culture. The results showed that the blood vessels formation by OCP/Gel group was significantly higher at 2 weeks than other groups but decreased at 4 weeks during the advancement of new bone formation. The capillary-like tube formation was highest in an OCP dose of 1 mg per well while other OCP doses above or below 1 mg did not show such a stimulatory effect. The results established both in vivo and in vitro confirmed that OCP has a positive effect on angiogenesis during bone regeneration in a suitable dose ranges, suggesting that the angiogenesis stimulated by OCP could be involved in the OCP/Gel-enhanced bone regeneration. STATEMENT OF SIGNIFICANCE: We have reported that octacalcium phosphate (OCP) materials display stimulatory capacities on the bone tissue-related cells. However, the effect of OCP on the angiogenesis and its relation to the OCP-enhanced bone regeneration is unknown. This study confirmed the capacity of OCP on angiogenesis before increasing the new bone mass after the implantation of a composite of OCP and gelatin (OCP/Gel). The blood vessels formation took place associated with the area beginning of the new bone formation, which finally decreased together with development of bone formation. Because OCP was ascertained stimulating the capillary-like tube formation in HUVEC culture with a certain OCP dose, the present study is the first report showing the capacity of OCP on angiogenesis during the OCP/Gel-enhanced bone regeneration.
Collapse
|
44
|
Nunes D, Ianus A, Shemesh N. Layer-specific connectivity revealed by diffusion-weighted functional MRI in the rat thalamocortical pathway. Neuroimage 2019; 184:646-657. [PMID: 30267858 PMCID: PMC6264401 DOI: 10.1016/j.neuroimage.2018.09.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
Investigating neural activity from a global brain perspective in-vivo has been in the domain of functional Magnetic Resonance Imaging (fMRI) over the past few decades. The intricate neurovascular couplings that govern fMRI's blood-oxygenation-level-dependent (BOLD) functional contrast are invaluable in mapping active brain regions, but they also entail significant limitations, such as non-specificity of the signal to active foci. Diffusion-weighted functional MRI (dfMRI) with relatively high diffusion-weighting strives to ameliorate this shortcoming as it offers functional contrasts more intimately linked with the underlying activity. Insofar, apart from somewhat smaller activation foci, dfMRI's contrasts have not been convincingly shown to offer significant advantages over BOLD-driven fMRI, and its activation maps relied on significant modelling. Here, we study whether dfMRI could offer a better representation of neural activity in the thalamocortical pathway compared to its (spin-echo (SE)) BOLD counterpart. Using high-end forepaw stimulation experiments in the rat at 9.4 T, and with significant sensitivity enhancements due to the use of cryocoils, we show for the first time that dfMRI signals exhibit layer specificity, and, additionally, display signals in areas devoid of SE-BOLD responses. We find that dfMRI signals in the thalamocortical pathway cohere with each other, namely, dfMRI signals in the ventral posterolateral (VPL) thalamic nucleus cohere specifically with layers IV and V in the somatosensory cortex. These activity patterns are much better correlated (compared with SE-BOLD signals) with literature-based electrophysiological recordings in the cortex as well as thalamus. All these findings suggest that dfMRI signals better represent the underlying neural activity in the pathway. In turn, these advanatages may have significant implications towards a much more specific and accurate mapping of neural activity in the global brain in-vivo.
Collapse
Affiliation(s)
- Daniel Nunes
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Andrada Ianus
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal; Centre for Medical Image Computing, University College London, London, UK
| | - Noam Shemesh
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
45
|
Abstract
We investigated the intraosseous arteries of six normal cadaveric scaphoids using red lead injection and three-dimensional reconstruction with micro-computed tomography. The arterial entrances were generally located around the dorsal ridge, the insertion of the scaphocapitate ligament and the radial part of the radioscaphocapitate ligament. Two to three trunk arteries entered the scaphoid on dorsal ridge at the level of the waist. The distal part of the scaphoid was mainly supplied by arteries from the waist. The blood supply of about 40% of proximal part of the scaphoid was poor. The blood supply from the scaphoid dorsal ridge plays an important role, not only for the proximal part of the scaphoid, but also for the waist and even the distal pole. The intrascaphoid arterial pattern may be contributory to nonunion, especially in proximal pole fractures.
Collapse
Affiliation(s)
- Zirun Xiao
- 1 Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, China.,2 Department of Orthopaedic Surgery, PLA 91st Central Hospital, Jiaozuo, China
| | - Ge Xiong
- 1 Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, China
| | - Weiguang Zhang
- 3 Department of Anatomy and Histology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
46
|
Simulations of blood as a suspension predicts a depth dependent hematocrit in the circulation throughout the cerebral cortex. PLoS Comput Biol 2018; 14:e1006549. [PMID: 30452440 PMCID: PMC6277127 DOI: 10.1371/journal.pcbi.1006549] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/03/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
Recent advances in modeling oxygen supply to cortical brain tissue have begun to elucidate the functional mechanisms of neurovascular coupling. While the principal mechanisms of blood flow regulation after neuronal firing are generally known, mechanistic hemodynamic simulations cannot yet pinpoint the exact spatial and temporal coordination between the network of arteries, arterioles, capillaries and veins for the entire brain. Because of the potential significance of blood flow and oxygen supply simulations for illuminating spatiotemporal regulation inside the cortical microanatomy, there is a need to create mathematical models of the entire cerebral circulation with realistic anatomical detail. Our hypothesis is that an anatomically accurate reconstruction of the cerebrocirculatory architecture will inform about possible regulatory mechanisms of the neurovascular interface. In this article, we introduce large-scale networks of the murine cerebral circulation spanning the Circle of Willis, main cerebral arteries connected to the pial network down to the microcirculation in the capillary bed. Several multiscale models were generated from state-of-the-art neuroimaging data. Using a vascular network construction algorithm, the entire circulation of the middle cerebral artery was synthesized. Blood flow simulations indicate a consistent trend of higher hematocrit in deeper cortical layers, while surface layers with shorter vascular path lengths seem to carry comparatively lower red blood cell (RBC) concentrations. Moreover, the variability of RBC flux decreases with cortical depth. These results support the notion that plasma skimming serves a self-regulating function for maintaining uniform oxygen perfusion to neurons irrespective of their location in the blood supply hierarchy. Our computations also demonstrate the practicality of simulating blood flow for large portions of the mouse brain with existing computer resources. The efficient simulation of blood flow throughout the entire middle cerebral artery (MCA) territory is a promising milestone towards the final aim of predicting blood flow patterns for the entire brain. The brain’s astonishing cognitive capacity depends on the coordination between neurons and the cerebral circulation, a system known as the neurovascular unit. The spatial and temporal coupling between these two networks is the object of intense research. However, the concise anatomical description of the cerebral circulation has so far been intractable. This paper introduces a methodology for the in silico creation of realistic models for the entire cerebral circulation. This innovation incorporates topological data from several neuroimaging modalities covering three lengths scales as input into a computer algorithm, which assembles anatomically accurate circulatory networks. When simulating blood flow as red blood cells suspended in plasma for experimental and synthetic cortical network models, we discovered that red blood cells tend to be more concentrated in deeper layers of the cortex compared to the surface. RBC fluxes are more homogenous in deeper layers. The phenomenon of depth dependent red blood cell supply supports the notion that the intricate architecture of the cortical microcirculation serves a self-regulating function to maintain uniform oxygen perfusion to neurons. We also demonstrate the practicality of predicting blood flow patterns for the entire brain with existing computer power.
Collapse
|
47
|
Xie L, Koukos G, Barck K, Foreman O, Lee WP, Brendza R, Eastham-Anderson J, McKenzie BS, Peterson A, Carano RAD. Micro-CT imaging and structural analysis of glomeruli in a model of Adriamycin-induced nephropathy. Am J Physiol Renal Physiol 2018; 316:F76-F89. [PMID: 30256127 DOI: 10.1152/ajprenal.00331.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glomeruli number and size are important for determining the pathogenesis of glomerular disease, chronic kidney disease, and hypertension. Moreover, renal injury can occur in specific cortical layers and alter glomerular spatial distribution. In this study, we present a comprehensive structural analysis of glomeruli in a model of Adriamycin (doxorubicin) nephropathy. Glomeruli are imaged (micro-CT at 10 × 10 × 10 μm3) in kidney specimens from C57Bl/6 mouse cohorts: control treated with saline ( n = 9) and Adriamycin treated with 20 mg/kg Adriamycin ( n = 7). Several indices were examined, including glomerular number, glomerular volume, glomerular volume heterogeneity, and spatial density at each glomerulus and in each cortical layer (superficial, midcortical, and juxtamedullary). In the Adriamycin-treated animals, glomerular number decreased significantly in the left kidney [control: 8,298 ± 221, Adriamycin: 6,781 ± 630 (mean ± SE)] and right kidney (control: 7,317 ± 367, Adriamycin: 5,522 ± 508), and glomerular volume heterogeneity increased significantly in the left kidney (control: 0.642 ± 0.015, Adriamycin: 0.786 ± 0.018) and right kidney (control: 0.739 ± 0.016, Adriamycin: 0.937 ± 0.023). Glomerular spatial density was not affected. Glomerular volume heterogeneity increased significantly in the superficial and midcortical layers of the Adriamycin cohort. Adriamycin did not affect glomerular volume or density metrics in the juxtamedullary region, suggesting a compensatory mechanism of juxtamedullary glomeruli to injury in the outer cortical layers. Left/right asymmetry was observed in kidney size and various glomeruli metrics. The methods presented here can be used to evaluate renal disease models with subtle changes in glomerular endowment locally or across the entire kidney, and they provide an imaging tool to investigate diverse interventions and therapeutic drugs.
Collapse
Affiliation(s)
- Luke Xie
- Biomedical Imaging, Genentech, South San Francisco, California
| | - Georgios Koukos
- Molecular Biology, Genentech, South San Francisco, California
| | - Kai Barck
- Biomedical Imaging, Genentech, South San Francisco, California
| | - Oded Foreman
- Pathology, Genentech, South San Francisco, California
| | - Wyne P Lee
- Translation Immunology, Genentech, South San Francisco, California
| | - Robert Brendza
- Neuroscience, Genentech, South San Francisco, California
| | | | - Brent S McKenzie
- Translation Immunology, Genentech, South San Francisco, California
| | - Andrew Peterson
- Molecular Biology, Genentech, South San Francisco, California
| | | |
Collapse
|
48
|
Hlushchuk R, Haberthür D, Djonov V. Ex vivo microangioCT: Advances in microvascular imaging. Vascul Pharmacol 2018; 112:2-7. [PMID: 30248380 DOI: 10.1016/j.vph.2018.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 10/28/2022]
Abstract
Therapeutic modulation of angiogenesis is believed to be a prospective powerful treatment strategy to modulate the microcirculation and therefore help millions of patients with cardiovascular and cancer diseases. The often-frustrating results from late-stage clinical studies indicate an urgent need for improved assessment of the pro- and anti-angiogenic compounds in preclinical stage of investigation. For such a proper assessment, detailed vascular visualization and adequate quantification are essential. Nowadays, there are few imaging modalities available, but none of them provides non-destructive 3D-visualization of the vasculature down to the capillary level. In many instances, the approaches cannot be combined with the subsequent histological or ultrastructural analysis. In this review, we address the latest developments in the microvascular imaging, namely, the microangioCT approach with a polymer-based contrast agent (μAngiofil). This approach allows time-efficient non-destructive 3D-imaging of the organ and its vasculature including the finest capillaries. Besides the superior visualization, the obtained detailed 3D information on the organ vasculature enables its 3D-skeletonization and further quantitative analysis. Probably the only significant limitation of the described approach is that it can be used only ex vivo, i.e., no longitudinal studies. In spite of this drawback, microangioCT with μAngiofil is a relatively simple and straightforward tool with a broad application range for studying physiological and pathological alterations in the microvasculature of any organ. It provides microvascular imaging at unprecedented level and enables correlative microscopy.
Collapse
|
49
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
50
|
Neulen A, Kosterhon M, Pantel T, Kirschner S, Goetz H, Brockmann MA, Kantelhardt SR, Thal SC. A Volumetric Method for Quantification of Cerebral Vasospasm in a Murine Model of Subarachnoid Hemorrhage. J Vis Exp 2018. [PMID: 30102288 PMCID: PMC6126573 DOI: 10.3791/57997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a subtype of hemorrhagic stroke. Cerebral vasospasm that occurs in the aftermath of the bleeding is an important factor determining patient outcome and is therefore frequently taken as a study endpoint. However, in small animal studies on SAH, quantification of cerebral vasospasm is a major challenge. Here, an ex vivo method is presented that allows quantification of volumes of entire vessel segments, which can be used as an objective measure to quantify cerebral vasospasm. In a first step, endovascular casting of the cerebral vasculature is performed using a radiopaque casting agent. Then, cross-sectional imaging data are acquired by micro computed tomography. The final step involves 3-dimensional reconstruction of the virtual vascular tree, followed by an algorithm to calculate center lines and volumes of the selected vessel segments. The method resulted in a highly accurate virtual reconstruction of the cerebrovascular tree shown by a diameter-based comparison of anatomical samples with their virtual reconstructions. Compared with vessel diameters alone, the vessel volumes highlight the differences between vasospastic and non-vasospastic vessels shown in a series of SAH and sham-operated mice.
Collapse
Affiliation(s)
- Axel Neulen
- Department of Neurosurgery, Medical Center of the Johannes Gutenberg - University;
| | - Michael Kosterhon
- Department of Neurosurgery, Medical Center of the Johannes Gutenberg - University
| | - Tobias Pantel
- Department of Neurosurgery, Medical Center of the Johannes Gutenberg - University
| | - Stefanie Kirschner
- Department of Neuroradiology, Medical Center of the Johannes Gutenberg - University
| | - Hermann Goetz
- Platform for Biomaterial Research, Medical Center of the Johannes Gutenberg - University
| | - Marc A Brockmann
- Department of Neuroradiology, Medical Center of the Johannes Gutenberg - University
| | - Sven R Kantelhardt
- Department of Neurosurgery, Medical Center of the Johannes Gutenberg - University
| | - Serge C Thal
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg - University;
| |
Collapse
|