Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Semi-automated protocol to quantify and characterize fluorescent three-dimensional vascular images

  • Danny F. Xie,

    Roles Data curation, Formal analysis, Investigation, Methodology, Software, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

  • Christian Crouzet,

    Roles Methodology, Supervision, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

  • Krystal LoPresti,

    Roles Conceptualization, Formal analysis, Investigation, Software, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

  • Yuke Wang,

    Roles Conceptualization, Investigation, Writing – original draft, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

  • Christopher Robinson,

    Roles Conceptualization, Investigation, Writing – original draft, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

  • William Jones,

    Roles Visualization, Writing – review & editing

    Affiliation Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America

  • Fjolla Muqolli,

    Roles Visualization, Writing – review & editing

    Affiliation Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America

  • Chuo Fang,

    Roles Investigation, Resources, Writing – review & editing

    Affiliation Department of Neurology, University of California-Irvine, Irvine, CA, United States of America

  • David H. Cribbs,

    Roles Funding acquisition, Resources, Writing – review & editing

    Affiliation Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, United States of America

  • Mark Fisher,

    Roles Funding acquisition, Resources, Writing – review & editing

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Neurology, University of California-Irvine, Irvine, CA, United States of America, Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, United States of America, Department of Pathology & Laboratory Medicine, University of California-Irvine, Irvine, CA, United States of America

  • Bernard Choi

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Project administration, Resources, Software, Supervision, Visualization, Writing – original draft, Writing – review & editing

    choib@uci.edu

    Affiliations Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America, Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America

Abstract

The microvasculature facilitates gas exchange, provides nutrients to cells, and regulates blood flow in response to stimuli. Vascular abnormalities are an indicator of pathology for various conditions, such as compromised vessel integrity in small vessel disease and angiogenesis in tumors. Traditional immunohistochemistry enables the visualization of tissue cross-sections containing exogenously labeled vasculature. Although this approach can be utilized to quantify vascular changes within small fields of view, it is not a practical way to study the vasculature on the scale of whole organs. Three-dimensional (3D) imaging presents a more appropriate method to visualize the vascular architecture in tissue. Here we describe the complete protocol that we use to characterize the vasculature of different organs in mice encompassing the methods to fluorescently label vessels, optically clear tissue, collect 3D vascular images, and quantify these vascular images with a semi-automated approach. To validate the automated segmentation of vascular images, one user manually segmented one hundred random regions of interest across different vascular images. The automated segmentation results had an average sensitivity of 83±11% and an average specificity of 91±6% when compared to manual segmentation. Applying this procedure of image analysis presents a method to reliably quantify and characterize vascular networks in a timely fashion. This procedure is also applicable to other methods of tissue clearing and vascular labels that generate 3D images of microvasculature.

Introduction

The microvasculature facilitates gas exchange, provides nutrients to cells, and regulates blood flow in response to stimuli [1]. Thus, it plays a fundamental role in the survival and health of tissues and organs. Vascular abnormalities indicate pathology for various conditions, such as compromised vessel integrity in cerebral microvascular disease and angiogenesis in tumors [24]. In addition, impaired blood flow to the brain is associated with neurodegenerative disorders such as Alzheimer’s disease [5, 6]. Traditional immunohistochemistry enables the visualization of tissue cross-sections containing exogenously labeled vasculature. Although this approach quantifies vascular changes within small fields of view, more practical ways exist to study the vasculature on the scale of whole organs. Furthermore, traditional immunohistochemistry requires tissue sectioning into thin (6–40 μm) sections, which effectively limits visualization of features to planar views and thus impedes facile 3D visualization of vascular architecture.

Volumetric imaging of tissue samples is vital to studying the microvasculature in its native state. The main limitation of 3D imaging of tissue samples is optical scattering resulting from the microscopic variations of refractive index occurring in most biological tissues. Organic and aqueous solvents reduce tissue turbidity by achieving refractive index matching [7, 8]. CLARITY is a popular tissue clearing method involving embedding of the sample into a hydrogel and using an electric current to remove lipids from the sample [9]. Other tissue clearing procedures have advantages and disadvantages associated with clearing time, the extent of clearing, and preservation of original tissue characteristics (e.g. size, endogenous fluorescence).

Quantitative analysis of microvasculature is essential to understand how structural variations in the microvascular network may change for different pathological states [10]. The feasibility of such analysis depends on accurate, ideally automated segmentation methods for isolating the microvasculature from the background due to the hundreds of gigabytes of data generated with high-resolution 3D imaging. Various groups have developed automated algorithms to perform quantitative characterization of vascular images [11, 12]. However, each method varies in complexity, processing time, and computational requirements.

We currently use iDISCO (immunolabeling-enabled three-dimensional imaging of solvent-cleared organs) as our primary tissue clearing method [7, 13]. Briefly, iDISCO consists of methanol dehydration, lipid removal with dichloromethane, and refractive index matching with dibenzyl ether. iDISCO is a fast, simple-to-implement clearing method that enables deep-tissue imaging. It is compatible with many exogenous labels often used for traditional immunohistochemistry. We previously demonstrated the effectiveness of iDISCO in combination with lectin-DyLight-649 for 3D visualization of vasculature in a mouse brain [13]. We also used Prussian blue labeling of hemosiderin [1416], a by-product of cerebral microhemorrhages, with iDISCO-cleared brains and lectin-DyLight-649 labeling.

In this paper, we first review different methods for vascular labeling in conjunction with optical clearing that other groups have published. Next, we describe our complete protocol for imaging the vasculature in different organs in mice (Fig 1A). Specifically, we report on sample collection and perfusion of lectin-DyLight-649 followed by adding additional labels as desired and optical clearing. We then describe our procedure to obtain 3D confocal microscopy images. Finally, we describe our semi-automated approach to process the resulting images and quantify the vascular architecture in three dimensions. The proposed methodology consists of simple procedures that require minimal computational resources, which enables other researchers to produce accurate three-dimensional vascular images.

thumbnail
Fig 1. Overview of protocol for vascular visualization and quantification with various examples published in literature.

A) Illustrations of the critical steps and workflow for vascular visualization in cleared tissue. B-D) Examples of vascular visualization in literature. B) Visualization of CD31-labeled mouse tumor vasculature with seeDB [30]. C) Visualization of DiI-labeled mouse heart vasculature with FocusClear [11]. D) Visualization of liver vasculature from Tie2-Cre mouse model with PEGASOS [33].

https://doi.org/10.1371/journal.pone.0289109.g001

Vascular imaging with various vessel labels and optical clearing techniques

Reviewing the literature on 3D vascular imaging of cleared tissue will show the large variability in methodology across different experiments (see Table 1 for specific references). For example, blood vessels can be visualized with transgenic animals expressing a fluorescent protein in endothelial cells, labeled in vivo with intravascular dyes, or labeled ex vivo with immersion-based antibody labeling. In addition, tissue preparation can vary from thick tissue sections and whole organs to whole-mount specimens, depending on the purpose of an experiment. Finally, clearing procedures can be grouped into aqueous solvents [1722], organic solvents [12, 2325], and hydrogel-based [2629].

thumbnail
Table 1. Summary of different approaches to visualizing 3D microvasculature in optically cleared tissues.

https://doi.org/10.1371/journal.pone.0289109.t001

When selecting a clearing procedure, it is essential to consider its compatibility with the chosen vascular label. Endogenous fluorescence intensity is known to be reduced when using the iDISCO protocol; hence, an alternative vascular label would be preferred. Imaging of cleared samples is typically performed with confocal microscopy or light-sheet microscopy. Lastly, vascular-related metrics, such as vessel diameters and vessel density, are quantified with existing software such as FIJI and Imaris or custom-designed software such as ClearMap 2.0.

We have reviewed several optical clearing methods utilized to image vasculature. They are organized in Table 1 by clearing method, tissue type, vessel label, and quantification. Vascular images in cleared tissue from select publications are included in Fig 1B–1D. Lee et al. (2017) demonstrated the ability to use seeDB, a water-based clearing method that relies on fructose, to visualize CD31-labeled endothelial cells in whole mouse tumors (Fig 1B, 1B’) [30, 31]. Our group visualized vessels with an intravascular fluorescent dye and FocusClear (Fig 1C, 1C’) [11]. In addition, we quantified functional vascular density in different regions of cardiac tissue with a custom MATLAB algorithm [11, 32]. With a polyethylene glycol-associated solvent system (PEGASOS), researchers developed a clearing technique that preserved both hard and soft tissue structures (Fig 1D, 1D’) [33], allowing for vascular imaging throughout an entire specimen.

Three-dimensional quantitative analysis of microvasculature imaged in thick tissue sections

Microvascular labeling is performed by injecting of a lectin conjugated to a fluorophore. In our work, we have focused on the DyLight-649 fluorophore. As the lectin travels through the circulatory system, it binds to glycoproteins adjacent to endothelial cells of the vascular wall. This binding allows for the labeling of the vascular network within every organ of the body.

An overview of our workflow is outlined below in Fig 2. First, we administer lectin-DyLight-649 via retroorbital injection. The lectin is allowed to circulate throughout the body before cardiac perfusion with saline, followed by formalin. Mouse brains are then extracted, bisected into hemispheres, and sectioned into thick (0.5–1.0mm) sections. Each section is then imaged using confocal microscopy to generate image stacks of the complete section throughout its entire thickness. Next, segmentation is performed by a custom MATLAB (MathWorks, Natick, MA) script. Finally, neuTube, an open-source neuron tracing software, is used to extract diameter measurements of the vessel structures [51].

thumbnail
Fig 2. Workflow for 3D visualization and quantitation of microvasculature in thick tissue sections.

A) Vasculature is labeled via retroorbital injection of lectin-DyLight-649. Brains are then sectioned and cleared. B) Depiction of multiplane imaging and image stitching to visualize an entire sample in 3D via confocal microscopy. C) From left to right: representative example of a raw image of the vasculature, segmented vasculature via MATLAB, 2D view of traced vasculature via neuTube, and 3D view of traced vasculature via neuTube.

https://doi.org/10.1371/journal.pone.0289109.g002

Materials and methods

The protocol described in this article is published on protocols.io (https://doi.org/10.17504/protocols.io.j8nlkk7m1l5r/v1) and is included as S1 File. All custom-written MATLAB code is available upon reasonable request made to the corresponding author.

Animals

This work was in compliance with protocols approved by the University of California, Irvine Institutional Animal Care and Use Committee (approval number: AUP-19-084, AUP-19-032). Anesthesia is induced at 4% isoflurane within a chamber and maintained at 1.5% isoflurane. Euthanasia is performed via cardiac perfusion under 4% isoflurane anesthesia.

Anticipated results

The procedures detailed herein present a simple method for producing optically cleared (> 1 mm thick) tissue samples with fluorescently labeled vasculature. Samples can then be imaged to generate 3D images of the microvasculature. The described segmentation method using custom MATLAB scripts and neuTube presents an automated method to convert the fluorescent images into binary images. The binary images can then be used to characterize microvasculature by quantifying vessel density and vessel sizes. The procedures here can be enhanced by using additional histology dyes or immunohistochemistry to label biomarkers or other structures of interest in tissue. In doing so, one can visualize and quantify the microvasculature surrounding these biomarkers to gain insight into their relationship.

Validation

Segmentation results

To validate the accuracy of our automatically segmented images, manually segmented images were created to serve as a ground truth comparison. One author (DFX) used MATLAB to manually outline every pixel within an ROI of an image that the author determined to belong to a vessel. Each ROI (n = 100) was presented as a maximum intensity projection (MIP) image of a 50x50x5 voxel (113x113x38 μm) region. A MIP is used to present a 2D image that is more feasible for manual tracing. The corresponding 50x50x5 voxel region MIP from the automated segmented image was compared. Four representative examples of this are depicted in Fig 3. Across the 100 ROIs, the average sensitivity was 83±11%, and the average specificity was 91±6%. The Dice similarity coefficient between manually segmented and automated segmented images was calculated across all ROIs. This coefficient can measure similarity between two sets of Boolean data and ranges between 0 and 1, where 1 represents identical data, and 0 represents opposite data [52]. The average Dice similarity coefficient across 100 ROIs was 0.74±0.09.

thumbnail
Fig 3. Representative examples of raw images, automated segmentation, and manual segmentation.

A) Automated segmentation is performed using the iterative selection method as described in the step-by-step protocol (see S1 File). Manual segmentation is performed by manual tracing of vessels. Each example shown is a 50x50 pixel region from a different brain section. Scale bar is 25 μm. B) Comparison of vessel pixels identified via manual segmentation (ground truth) and automated segmentation. The sensitivity, specificity, and Dice similarity coefficient of all 100 ROIs are shown in the graph. The average sensitivity was 83±11%, the average specificity was 91±6%, and the average Dice similarity coefficient was 0.74±0.09.

https://doi.org/10.1371/journal.pone.0289109.g003

Diameter results with neuTube

Manually measured diameters of individual vessels were used as a ground truth comparison for neuTube approximated vessel diameters. Images from various brain sections across three different animals were used. One author (DFX) was presented with a randomly generated 50x50x5 voxel (113x113x38 μm) size region as a MIP. Within the image, the user was tasked with using MATLAB to estimate the diameter of a single vessel in the image at five different points along the vessel by drawing lines approximately perpendicular to the vessel’s centerline. In neuTube, five nodes along the corresponding vessel were selected. The average of the five manual diameter measurements via MATLAB and the five automated diameter measurements via neuTube were compared. Two representative examples of this are depicted in Fig 4A. The absolute difference between diameter measurements was 1.16±0.73 μm as shown in Fig 4B (n = 50 vessels of diameters ranging from 1.78 μm to 3.19 μm).

thumbnail
Fig 4. Two representative examples of manual diameter measurements and neuTube diameter measurements.

A) Within a 50x50 pixel region, one vessel is chosen by the user for analysis. The manual diameter measurement is determined by drawing a line approximately perpendicular to the centerline of the vessel at 5 locations (shown with red lines in the top row of images). The manual diameter measurement for a given vessel is the average of these 5 measurements. The same vessel is identified within the automated neuTube output where each node (shown as a sphere in the bottom row of images) represents the diameter determined by neuTube at that location in the image. The neuTube diameter measurement for a given vessel is recorded as the average of five nodes along that vessel Scale bar is 25 μm. B) The absolute difference in diameter between manual measurement and neuTube measurement for 50 different vessels is shown. The mean difference was 1.16±0.73 μm.

https://doi.org/10.1371/journal.pone.0289109.g004

Discussion

Due to the understood importance of cerebrovascular architecture to brain function, several groups have reported on similar methods to what we report here. Lugo-Hernandez et al. (2017) use an injected fluorescent-labeled hydrogel, the 3DISCO optical clearing approach, and light-sheet microscopy to image the vasculature, but vascular analysis is performed using a commercial software package (Imaris), which uses both user intervention and proprietary algorithms to achieve binary vascular maps [24]. Di Giovanna et al. (2018) employed a fluorescent gel perfusion approach to fill the vasculature, CLARITY-based optical clearing, and light-sheet microscopy; they describe the use of an automatic segmentation method based on a Markov random field, but enabling details of the algorithm are not provided [26]. Quintana et al. (2019) and Wälchli et al. (2021) used a vascular corrosion cast approach and micro-computerized tomography to visualize the whole-brain vasculature with exquisite detail, along with automated thresholding approaches (including the same iterative selection method we describe here); however, this approach requires access to a micro-CT system, and its compatibility with fluorescence labels requires further investigation [53, 54]. Kirst et al. (2020) report on the combination of immunolabeling, iDISCO-based optical clearing, and light-sheet microscopy to achieve whole-brain three-dimensional maps of the cerebrovasculature. They provide a sophisticated open-source software package to create binary maps of the vasculature, but a high-performance dedicated workstation is required to execute their computationally-intensive approach [12]. Hahn et al. (2021) used the same lectin-Dylight-649 fluorophore as we describe here, along with the FluoClearBABB optical clearing approach and light-sheet microscope; they describe an automated image analysis approach using a trained random forest classification scheme “ilastik” (Berg et al. 2019) [55, 56]. Takahashi et al. (2022) used the Cre/lox approach to induce Td tomato expression in endothelial cells, and combined this with CUBIC-based optical clearing and light-sheet microscopy to image the vasculature; they too used the ilastik software module for analysis [57]. Zhu et al. (2022) used different combinations of vascular labels and optical clearing approaches with light-sheet microscopy, but they also used Imaris for vascular image analysis [58].

With the work described here, we focused on describing the entire process of vessel labeling, brain extraction, and quantitative image analysis, on enabling other researchers to integrate and expand upon our protocol into their individualized workflow. We acknowledge that other vessel segmentation approaches may offer advantages to our approach described here, and the vetting of those approaches will be the subject of future work. Here, we emphasize the transparency of our methodology and ease of implementation of each step, with minimal computational resources, to achieve an approach that can rapidly yield accurate three-dimensional vascular maps. The vessel segmentation approach readily works with similarly-prepared whole brains imaged with light-sheet microscopy [13].

There are several potential applications for the methods described here. As angiogenesis occurs in organs, endothelial cells respond to local signals to adapt vessels to the surrounding environment [59]. Angiogenesis plays an essential role in tumor formation. In tumors, angiogenesis directly impacts tumor growth and metastasis. Overexpression of proangiogenic factors leads to uncontrolled vascular growth in tumors [60]. As a result, anti-angiogenic drugs are frequently used as a potential treatment option for cancer. Lectin-DyLight-649 combined with iDISCO presents a robust procedure for labeling the microvasculature in all body areas, including tumors. Changes in vessel density, tortuosity, and diameters can be quantified to evaluate the efficacy of novel anti-angiogenic treatments.

Ischemic strokes occur when there is a significant drop in cerebral blood flow due to occlusion in a cerebral artery. A major consequence of such an event is necrosis of neurons due to a deficient blood supply [61]. A method to visualize the microvascular network and the surrounding neurons, astrocytes, and glial cells can provide a detailed 3D snapshot of the brain in response to an ischemic stroke and monitor potential treatments over time.

In addition to visualizing cerebral microhemorrhages within the surrounding microvascular network and providing the improved capability to estimate the size range of these lesions, our approach can provide enhanced imaging for other disease entities. For example, a growing body of literature suggests a contribution of dysfunctional regulation of cerebral blood flow and various types of cognitive impairment [5]. The ability to visualize the cerebral microvasculature in three dimensions offers a new gateway, potentially leading to the identification of novel treatment targets for neurological disorders.

Supporting information

Acknowledgments

This study was made possible in part through access to the Optical Biology Core Facility of the Developmental Biology Center, a shared resource supported by the Cancer Center Support Grant (CA-62203) and Center for Complex Biological Systems Support Grant (GM-076516) at the University of California, Irvine.

References

  1. 1. Jacob M, Chappell D, Becker BF. Regulation of blood flow and volume exchange across the microcirculation. Crit Care. 2016;20(1):1–13. Available from: pmid:27765054
  2. 2. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011 May 18;473(7347):298–307. Available from: pmid:21593862
  3. 3. Haller S, Vernooij MW, Kuijer JPA, Larsson EM, Jäger HR, Barkhof F. Cerebral Microbleeds: Imaging and Clinical Significance. Radiology. 2018 Apr;287(1):11–28. Available from: pmid:29558307
  4. 4. Fang C, Magaki SD, Kim RC, Kalaria RN, Vinters H V., Fisher M. Arteriolar neuropathology in cerebral microvascular disease. Neuropathol Appl Neurobiol. 2023 Feb;49(1):1–18. Available from: pmid:36564356
  5. 5. Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic B V. The role of brain vasculature in neurodegenerative disorders. Nat Neurosci. 2018;21(10):1318–31. Available from: pmid:30250261
  6. 6. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77(9):1745–70. Available from: pmid:31690961
  7. 7. Renier N, Wu Z, Simon DJ, Yang J, Ariel P, Tessier-Lavigne M. IDISCO: A simple, rapid method to immunolabel large tissue samples for volume imaging. Cell. 2014;159(4):896–910. Available from: pmid:25417164
  8. 8. Susaki EA, Tainaka K, Perrin D, Kishino F, Tawara T, Watanabe TM, et al. Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis. Cell. 2014;157(3):726–39. Available from: pmid:24746791
  9. 9. Chung K, Wallace J, Kim SY, Kalyanasundaram S, Andalman AS, Davidson TJ, et al. Structural and molecular interrogation of intact biological systems. Nature. 2013 May 16;497(7449):332–7. Available from: pmid:23575631
  10. 10. Kalaria RN, Akinyemi R, Ihara M. Stroke injury, cognitive impairment and vascular dementia. Biochim Biophys Acta—Mol Basis Dis. 2016 May;1862(5):915–25. Available from: pmid:26806700
  11. 11. Moy AJ, Lo PC, Choi B. High-resolution visualization of mouse cardiac microvasculature using optical histology. Biomed Opt Express. 2014 Jan 1;5(1):69. Available from: pmid:24466477
  12. 12. Kirst C, Skriabine S, Vieites-Prado A, Topilko T, Bertin P, Gerschenfeld G, et al. Mapping the Fine-Scale Organization and Plasticity of the Brain Vasculature. Cell. 2020;180(4):780–795.e25. Available from: pmid:32059781
  13. 13. Khouri K, Xie DF, Crouzet C, Bahani AW, Cribbs DH, Fisher MJ, et al. Simple methodology to visualize whole-brain microvasculature in three dimensions. Neurophotonics. 2021 Apr 19;8(02):1–10. Available from: pmid:33884280
  14. 14. Crouzet C, Jeong G, Chae RH, LoPresti KT, Dunn CE, Xie DF, et al. Spectroscopic and deep learning-based approaches to identify and quantify cerebral microhemorrhages. Sci Rep. 2021 Dec 21;11(1):10725. Available from: pmid:34021170
  15. 15. Lo P, Crouzet C, Vasilevko V, Choi B. Visualization of microbleeds with optical histology in mouse model of cerebral amyloid angiopathy. Microvasc Res. 2016;105:109–13. Available from: pmid:26876114
  16. 16. Sumbria RK, Grigoryan MM, Vasilevko V, Paganini-Hill A, Kilday K, Kim R, et al. Aging exacerbates development of cerebral microbleeds in a mouse model. J Neuroinflammation. 2018 Dec 6;15(1):69. Available from: pmid:29510725
  17. 17. Zhu J, Yu T, Li Y, Xu J, Qi Y, Yao Y, et al. MACS: Rapid Aqueous Clearing System for 3D Mapping of Intact Organs. Adv Sci. 2020 Apr 25;7(8):1903185. Available from: pmid:32328422
  18. 18. Hama H, Hioki H, Namiki K, Hoshida T, Kurokawa H, Ishidate F, et al. ScaleS: an optical clearing palette for biological imaging. Nat Neurosci. 2015 Oct 14;18(10):1518–29. Available from: pmid:26368944
  19. 19. Nehrhoff I, Ripoll J, Samaniego R, Desco M, Gómez-Gaviro MV. Looking inside the heart: a see-through view of the vascular tree. Biomed Opt Express. 2017 Jun 1;8(6):3110. Available from: pmid:28663930
  20. 20. Nishimura W, Sakaue-Sawano A, Takahashi S, Miyawaki A, Yasuda K, Noda Y. Optical clearing of the pancreas for visualization of mature β-cells and vessels in mice. Islets. 2018 May 4;10(3):e1451282. Available from: pmid:29617192
  21. 21. Fu YY, Tang SC. Optical clearing facilitates integrated 3D visualization of mouse ileal microstructure and vascular network with high definition. Microvasc Res. 2010;80(3):512–21. Available from: pmid:20600164
  22. 22. Loren M, Crouzet C, Bahani A, Vasilevko V, Choi B. Optical clearing potential of immersion-based agents applied to thick mouse brain sections. PLoS One. 2019 May 10;14(5):e0216064. Available from: pmid:31075111
  23. 23. Qi Y, Yu T, Xu J, Wan P, Ma Y, Zhu J, et al. FDISCO: Advanced solvent-based clearing method for imaging whole organs. Sci Adv. 2019 Jan 4;5(1):1–14. Available from: pmid:30746463
  24. 24. Lugo-Hernandez E, Squire A, Hagemann N, Brenzel A, Sardari M, Schlechter J, et al. 3D visualization and quantification of microvessels in the whole ischemic mouse brain using solvent-based clearing and light sheet microscopy. J Cereb Blood Flow Metab. 2017 Oct 28;37(10):3355–67. Available from: pmid:28350253
  25. 25. Cai R, Pan C, Ghasemigharagoz A, Todorov MI, Förstera B, Zhao S, et al. Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull–meninges connections. Nat Neurosci. 2019;22(2):317–27. Available from: pmid:30598527
  26. 26. Di Giovanna AP, Tibo A, Silvestri L, Müllenbroich MC, Costantini I, Allegra Mascaro AL, et al. Whole-Brain Vasculature Reconstruction at the Single Capillary Level. Sci Rep. 2018 Aug 22;8(1):12573. Available from: pmid:30135559
  27. 27. Krolewski DM, Kumar V, Martin B, Tomer R, Deisseroth K, Myers RM, et al. Quantitative validation of immunofluorescence and lectin staining using reduced CLARITY acrylamide formulations. Brain Struct Funct. 2018 Mar 14;223(2):987–99. Available from: pmid:29243106
  28. 28. Carrillo M, Chuecos M, Gandhi K, Bednov A, Moore DL, Maher J, et al. Optical tissue clearing in combination with perfusion and immunofluorescence for placental vascular imaging. Medicine (Baltimore). 2018 Sep;97(39):e12392. Available from: pmid:30278515
  29. 29. Singh JN, Nowlin TM, Seedorf GJ, Abman SH, Shepherd DP. Quantifying three-dimensional rodent retina vascular development using optical tissue clearing and light-sheet microscopy. J Biomed Opt. 2017 Jul 18;22(07):1. Available from: pmid:28717817
  30. 30. Lee SS young Bindokas VP, Kron SJ. Multiplex three-dimensional optical mapping of tumor immune microenvironment. Sci Rep. 2017;(November):1–11. Available from: pmid:29208908
  31. 31. Ke M tsen, Fujimoto S, Imai T. SeeDB: a simple and morphology-preserving optical clearing agent for neuronal circuit reconstruction. Nat Neurosci. 2013 Aug 23;16(8):1154–61. Available from: pmid:23792946
  32. 32. White SM, George SC, Choi B. Automated computation of functional vascular density using laser speckle imaging in a rodent window chamber model. Microvasc Res. 2011;82(1):92–5. Available from: pmid:21419785
  33. 33. Jing D, Zhang S, Luo W, Gao X, Men Y, Ma C, et al. Tissue clearing of both hard and soft tissue organs with the pegasos method. Cell Res. 2018;28(8):803–18. Available from: pmid:29844583
  34. 34. Matryba P, Sosnowska A, Wolny A, Bozycki L, Greig A, Grzybowski J, et al. Systematic Evaluation of Chemically Distinct Tissue Optical Clearing Techniques in Murine Lymph Nodes. J Immunol. 2020 Mar 1;204(5):1395–407. Available from: pmid:31953352
  35. 35. Miyawaki T, Morikawa S, Susaki EA, Nakashima A, Takeuchi H, Yamaguchi S, et al. Visualization and molecular characterization of whole-brain vascular networks with capillary resolution. Nat Commun. 2020;11(1):1–11. Available from: pmid:32107377
  36. 36. McKey J, Cameron LA, Lewis D, Batchvarov IS, Capel B. Combined iDISCO and CUBIC tissue clearing and lightsheet microscopy for in toto analysis of the adult mouse ovary†. Biol Reprod. 2020 Apr 24;102(5):1080–9. Available from: pmid:31965156
  37. 37. Zhu D, Wang J, Zhi Z, Wen X, Luo Q. Imaging dermal blood flow through the intact rat skin with an optical clearing method. J Biomed Opt. 2010;15(2):026008. Available from: pmid:20459253
  38. 38. Liebmann T, Renier N, Bettayeb K, Greengard P, Tessier-Lavigne M, Flajolet M. Three-Dimensional Study of Alzheimer’s Disease Hallmarks Using the iDISCO Clearing Method. Cell Rep. 2016;16(4):1138–52. Available from: pmid:27425620
  39. 39. Khoradmehr A, Mazaheri F, Anvari M, Tamadon A. A Simple Technique for Three-Dimensional Imaging and Segmentation of Brain Vasculature U sing Fast Free-of-Acrylamide Clearing Tissue in Murine. Cell J. 2019 Apr;21(1):49–56. Available from: pmid:30507088
  40. 40. Ivins S, Roberts C, Vernay B, Scambler PJ. Analysis of Coronary Vessels in Cleared Embryonic Hearts. J Vis Exp. 2016 Dec 7;2016(118):1–9. Available from: pmid:28060348
  41. 41. Kolesová H, Čapek M, Radochová B, Janáček J, Sedmera D. Comparison of different tissue clearing methods and 3D imaging techniques for visualization of GFP-expressing mouse embryos and embryonic hearts. Histochem Cell Biol. 2016 Aug 4;146(2):141–52. Available from: pmid:27145961
  42. 42. Harrison CH, Buckland GR, Brooks SE, Johnston DA, Chatelet DS, Liu AKL, et al. A novel method to visualise the three-dimensional organisation of the human cerebral cortical vasculature. J Anat. 2018 Jun;232(6):1025–30. Available from: pmid:29520782
  43. 43. Oren R, Fellus-Alyagor L, Addadi Y, Bochner F, Gutman H, Blumenreich S, et al. Whole Organ Blood and Lymphatic Vessels Imaging (WOBLI). Sci Rep. 2018;8(1):1–9. Available from: pmid:29362484
  44. 44. Huang J, Brenna C, Khan A ul M, Daniele C, Rudolf R, Heuveline V, et al. A cationic near infrared fluorescent agent and ethyl-cinnamate tissue clearing protocol for vascular staining and imaging. Sci Rep. 2019 Dec 24;9(1):521. Available from: pmid:30679514
  45. 45. Lagerweij T, Dusoswa SA, Negrean A, Hendrikx EML, de Vries HE, Kole J, et al. Optical clearing and fluorescence deep-tissue imaging for 3D quantitative analysis of the brain tumor microenvironment. Angiogenesis. 2017 Nov 11;20(4):533–46. Available from: pmid:28699046
  46. 46. Yi Y, Men Y, Jing D, Luo W, Zhang S, Feng JQ, et al. 3‐dimensional visualization of implant‐tissue interface with the polyethylene glycol associated solvent system tissue clearing method. Cell Prolif. 2019 May 3;52(3):e12578. Available from: pmid:30714253
  47. 47. Li Y, Song Y, Zhao L, Gaidosh G, Laties AM, Wen R. Direct labeling and visualization of blood vessels with lipophilic carbocyanine dye DiI. Nat Protoc. 2008 Nov 9;3(11):1703–8. Available from: pmid:18846097
  48. 48. Peviani M, Spano G, Pagani A, Brugnara G, Covino C, Galli R, et al. Lipophilic dye-compatible brain clearing technique allowing correlative magnetic resonance/high-resolution fluorescence imaging in rat models of glioblastoma. Sci Rep. 2020;10(1):1–15. Available from: pmid:33087842
  49. 49. Moy AJ, Capulong B V., Saager RB, Wiersma MP, Lo PC, Durkin AJ, et al. Optical properties of mouse brain tissue after optical clearing with FocusClearTM. J Biomed Opt. 2015 Sep 21;20(9):095010. Available from: pmid:26388460
  50. 50. Moy AJ, Wiersma MP, Choi B. Optical Histology: A Method to Visualize Microvasculature in Thick Tissue Sections of Mouse Brain. PLoS One. 2013 Jan 23;8(1):e53753. Available from: pmid:23372668
  51. 51. Feng L, Zhao T, Kim J. neuTube 1.0: A New Design for Efficient Neuron Reconstruction Software Based on the SWC Format. eneuro. 2015 Jan 29;2(1):ENEURO.0049-14.2014. Available from: pmid:26464967
  52. 52. Dice LR. Measures of the Amount of Ecologic Association Between Species. Ecology. 1945;26(3):297–302.
  53. 53. Quintana DD, Lewis SE, Anantula Y, Garcia JA, Sarkar SN, Cavendish JZ, et al. NeuroImage The cerebral angiome: High resolution MicroCT imaging of the whole brain cerebrovasculature in female and male mice. Neuroimage. 2019;202(April):116109. Available from: pmid:31446129
  54. 54. Wälchli T, Bisschop J, Miettinen A, Ulmann-schuler A, Hintermüller C, Meyer EP, et al. Hierarchical imaging and computational analysis of three-dimensional vascular network architecture in the entire postnatal and adult mouse brain. Nat Protoc. 2021;16(October). Available from: pmid:34480130
  55. 55. Hahn A, Bode J, Alexander A, Karimian-jazi K, Schregel K, Schwarz D, et al. Large-scale characterization of the microvascular geometry in development and disease by tissue clearing and quantitative ultramicroscopy. J Cereb Blood Flow Metab. 2021;41(7):1536–46. Available from: pmid:33043767
  56. 56. Berg S, Kutra D, Kroeger T, Straehle CN, Kausler BX, Haubold C, et al. ilastik: interactive machine learning for (bio) image analysis. Nat Methods. 2019;16(December). Available from: pmid:31570887
  57. 57. Takahashi K, Abe K, Kubota SI, Fukatsu N, Morishita Y, Yoshimatsu Y, et al. An analysis modality for vascular structures combining tissue-clearing technology and topological data analysis. Nat Commun. 2022;13(1):5239. Available from: pmid:36097010
  58. 58. Zhu J, Deng Y, Yu T, Liu X, Li D, Zhu D. Optimal combinations of fluorescent vessel labeling and tissue clearing methods for three-dimensional visualization of vasculature. Neurophotonics. 2022;9(4). Available from: pmid:36466188
  59. 59. McDonald DM. Angiogenesis and Vascular Remodeling in Inflammation and Cancer: Biology and Architecture of the Vasculature. In: Angiogenesis. Boston, MA: Springer US; 2008. p. 17–33. Available from: https://doi.org/10.1007/978-0-387-71518-6_2
  60. 60. Li T, Kang G, Wang T, Huang H. Tumor angiogenesis and anti‑angiogenic gene therapy for cancer (Review). Oncol Lett. 2018 May 17;16(1):687–702. Available from: pmid:29963134
  61. 61. Datta A, Sarmah D, Mounica L, Kaur H, Kesharwani R, Verma G, et al. Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl Stroke Res. 2020 Dec 26;11(6):1185–202. Available from: pmid:32219729