Abstract
Microvascular networks play key roles in oxygen transport and nutrient delivery to meet the varied and dynamic metabolic needs of different tissues throughout the body, and their spatial architectures of interconnected blood vessel segments are highly complex. Moreover, functional adaptations of the microcirculation enabled by structural adaptations in microvascular network architecture are required for development, wound healing, and often invoked in disease conditions, including the top eight causes of death in the Unites States. Effective characterization of microvascular network architectures is not only limited by the available techniques to visualize microvessels but also reliant on the available quantitative metrics that accurately delineate between spatial patterns in altered networks. In this review, we survey models used for studying the microvasculature, methods to label and image microvessels, and the metrics and software packages used to quantify microvascular networks. These programs have provided researchers with invaluable tools, yet we estimate that they have collectively attained low adoption rates, possibly due to limitations with basic validation, segmentation performance, and nonstandard sets of quantification metrics. To address these existing constraints, we discuss opportunities to improve effectiveness, rigor, and reproducibility of microvascular network quantification to better serve the current and future needs of microvascular research.
Keywords: blood vessels, image analysis, image quantification, microvasculature, vascular network, vessel architecture
Abbreviations
- 3D
three‐dimensional
- ASMA
alpha smooth muscle actin
- CD31
cluster of differentiation 31
- CD34
cluster of differentiation 34
- Col‐IV
collagen‐IV
- CRISPR
clustered regularly‐interspaced short palindromic repeats
- DAPI
4′,6‐diamidino‐2‐phenylindole
- DsRed
a red fluorescent protein
- EGFP
enhanced green fluorescent protein
- EMCN
endomucin
- EP
endpoints
- FLK‐1
fetal liver kinase‐1
- FLT‐1
FMS‐like tyrosine kinase‐1
- FPALM
photo‐activated localization microscopy
- GFP
green fluorescent protein
- HSC
hematopoietic stem cells
- IB4
griffonia simplicifolia lectin I isolectin B4
- MSC
mesenchymal stem cells
- NG2
neural/glial antigen 2
- OCT
optical coherence tomography
- PDGFRβ
platelet‐derived growth factor receptor beta
- PECAM
platelet endothelial cell adhesion molecule (CD31)
- RAVE
rapid analysis of vessel elements
- RFP
red fluorescent protein (eg, DsRed)
- SSR
sum of squared residuals
- Tie1
tyrosine kinase with immunoglobulin‐like and EGF‐like homology
- Tie2
angiopoietin‐1 receptor
- VAF
vessel area fraction
- VE‐cadherin/VE‐cad
vascular endothelial cadherin
- VEGFR
vascular endothelial growth factor receptor
- vWf
Von Willebrand factor
1. INTRODUCTION
The microvasculature plays a plethora of key roles in maintaining tissue homeostasis, including modulating oxygen transport,1 nutrient delivery, inflammation response,2 and wound healing.3 Structural changes to the microvascular architecture have been shown to profoundly regulate these fundamental biologic processes.4 Therefore, characterization of the complex changes in spatial structure of the microvascular architecture gives a better understanding of the roles microvessels play in pathogenesis, maintenance, prevention, and amelioration of diseases. Indeed, the importance of the microvasculature has long been appreciated in diseases such as small vessel disease,5 coronary microvascular disease,6 and the abundance of complications associated with diabetes.7 However, recent research has indicated that the microvasculature also plays key roles in the top eight causes of death in the United States8 (Figure 1) and many others, including (1) heart disease: impaired infarct wound healing,9 reduced oxygenation,10 pulmonary hypertension in pre‐capillary and post‐capillary vessels11; (2) cancer: pathological angiogenesis,12 enriched microvessel permeability,13 significant route for metastasis14; (3) lower respiratory disease: capillary dropout,15 reduced muscle oxygenation,16 airway rigidity from vasodilation17; (4) stroke: impaired microvascular flow patterns18 and reduced oxygenation,19 pericyte constriction of capillaries,20 dropout of functioning capillaries21; (5) unintentional injuries: angiogenesis,22 clot formation,23 immune cell recruitment24; (6) Alzheimer's: attenuated vasodilation response,25 amyloid angiopathy,26 and tissue hypoxia25; (7) diabetes mellitus: capillary permeability, pericyte dropout, capillary dropout27; and (8) pneumonia and influenzas: capillary permability,28 immune cell recruitment,28 and impaired lung oxygen transport.29 Additionally, the microvasculature is recognized as one of the most promising routes of drug delivery30 by enabling direct targeting of microvascular endothelial cells with intravascularly injected drugs to exert profound therapeutic effects in disease conditions.31 The overall import of the microvasculature in biomedical research is quickly approaching that of the nearly ubiquitous roles that the immune system plays in basic organismal processes32, 33 and disease development,34 and future research focused on microvascular structure, function, and adaptations promises profound opportunities for curing human disease.
Figure 1.
The significance of the microvasculature in top causes of death and disease in United States. Top eight classes of fatal disease or injury with the fraction of annual deaths in the United States. Included with each malady are three highlighted fundamental roles the microvasculature plays with initiation, maintenance, or treatment (see main text for references)
In this review, we highlight new key developments and survey contemporary and classical models of the microvasculature, along with techniques to label and image microvessels at high resolution where the complete microvascular structure is captured. Therefore, microvascular research that fails to resolve the smallest‐sized vascular structures is omitted or given less emphasis, such as fundus imaging of the retina35 and other clinical imaging methods. Although a subset of the modalities covered can yield 3D images, we focus on analysis of 2D projections of 3D vessel networks since it can be universally applied to all microvascular imaging modalities, 2D representations of 3D networks retain much of their information,36 and 2D methods for quantification of vessel architecture can be extended to three dimensions,37 although we do comment on the potential pitfalls of using 2D metrics to characterize 3D microvascular structures. Furthermore, many of the 3D modalities for microvascular imaging have reduced axial resolution compared to lateral, and practical considerations of acquisition time usually lead to further reduced axial sampling.38 The currently available programs to analyze and quantify microvascular structures are also covered, along with constructive proposals for improvement in this area. While each topic covered could be a detailed examination on its own, this review is meant to offer a basic orientation of the technological options available for microvascular research and a perspective on analytical techniques to increase scientific rigor as science faces an ongoing crisis in reproducibility.39
2. MARKERS AND MODELS TO STUDY MICROVASCULAR NETWORK ARCHITECTURE
The study of the complex architecture of microvasculature requires proper labeling and visualization of microvessels, using either a marker for particular cell types, unique basement membrane constituents, and/or labeling perfused microvessels via the intravascular injection of a dye or fluorescently tagged antibody to visualize blood flow through microvessel lumens.40, 41, 42 All of these labeling methods provide a means of contrasting microvascular architectures with the surrounding tissue when paired with a suitable imaging modality. The particular choice of vascular marker and imaging method should be carefully evaluated based on the biological questions being examined and determined based on requirements for resolution, signal to noise, tissue penetration depth, imaging location in terms of in vivo/ex vivo, and labeled cell specificity.38 The relative importance of the various labeling and imaging considerations for microvascular visualization depends on the nature of the research and biological questions asked. For example, with investigations focusing on angiogenesis and subsets of vessel types, cell labeling of specific subpopulations is essential, while for studies characterizing blood flow, accurate vessel diameter and connectivity between vessel segments have greater significance. Moreover, effective pairing of these technologies with a particular imaging approach requires an understanding of the fundamental strengths and weaknesses of the options available.
2.1. Markers of microvasculature
A critically important aspect of studying blood vessels is carefully tailoring biological interpretations and conclusions to appropriately correspond to the specific cell types or structures visualized (Table 1). An example that illustrates incongruity between data and interpretation is when vessels are labeled via perfusion of fluorescent dye43 and general conclusions are made about vascular remodeling, disregarding changes in structure of nonperfused vessels, vessel neosprouts, and regressing vessels.44 Even with specifically worded conclusions, focusing on findings that only quantify portions of the microvascular architecture represents an incomplete analysis and may omit significant remodeling events. Another key example is the use of superfused IB4 lectin, a marker commonly used to label blood vessels that also labels pericyte45 and macrophages46 (Figure 2A). Especially in development, many papers prematurely conclude changes occur in vascular architecture based on lectin staining47, 48 while omitting the issue that a mix of cell types are labeled, especially with the inability to differentiate structures between pericyte and endothelial cells. Studies that use Col‐IV staining to quantify microvascular architecture49 have similar shortcomings, labeling not just blood vessels50 (Figure 2B), but other cell types such as pericytes and fibroblasts.51 Additionally, Col‐IV also marks thin bridges between capillaries previously referred to as Col‐IV sleeves,52 string vessels,53 and acellular capillaries54 in various tissues such as retina,54 brain,55 and muscle. Especially in the retina, this feature is interpreted as a sign of collapsed or regressed vessels, yet this has never actually been established. There is a clear separation between the two structures in thickness (Figure 2C) and with the cross‐sectional pixel intensity profile between lumenized vessels and Col‐IV tracks (Figure 2D), with a lack of structures found in an intermediate or transitioning phenotype. An alternative hypothesis would be pericytes extending off‐vessel processes56 and secreting Col‐IV.57 For instances where the cell type responsible for an immunostained structure is not established with confidence, we caution interpreting results are cell‐type specific, even if previously stated in the literature.
Table 1.
Markers of the Microvasculature
Diagram |
![]() |
![]() |
![]() |
![]() |
|
---|---|---|---|---|---|
Name | Type | endothelial cells | pericytes | smooth muscle cells | Other |
PECAM/ CD31 | Surface136 | ✓137 | ✗138 | ✗139 | Platelets, T‐cells. leukocytes137 |
IB4 Lectin | Var.140, 141 | ✓46, 142 | ✓45 | ✓143 | Macrophages144, microglia46, monocytes145 |
Col‐IV | ECM40 | ✓146 | ✓147 | ✓148 | Fibroblasts149, MSCs150 |
Laminin | ECM151 | ✓152 | ✓152 | ✓152 | Fibroblasts153, epithelial cells154 |
CD34 | Surface136 | ✓155 | ✗156 | ✗157 | MSCs, HSCs, muscle satellite cells, fibrocytes158 |
VE‐cadherin | Surface138 | ✓138 | ✗159 | ✗160 | None161 |
EMCN | Surface162 | ✓163 | ✗164 | ✗164 | Putative hematopoietic progenitor cells165 |
vWf | Internal166 | ✓167 | ✗167 | ✗168 | Megakaryocytes166 |
aSMA | Internal169 | ✗169 | ✓170 | ✓171 | Fibroblasts172, perisinusoidal cells169 |
MYH11 | Internal173 | ✗173 | ✓174 | ✓175 | None174 |
NG2 | Surface176 | ✗176 | ✓157 | ✓156 | Microglia170, macrophages, oligodendrocyte porogenitors177 |
N‐Cadherin | Surface178 | ✓179 | ✓159 | ✓180 | MSCs181, fibroblasts159, osteoblasts159 |
Desmin | Internal182 | ✗168 | ✓183 | ✓156 | Interstitial cells, muscle satellite cells158 |
PDGFRβ | Surface184 | ✗185 | ✓156 | ✓156 | MSCs156, fibroblasts156 , some neurons156 |
Perf. PECAM | Surface186, 187 | Perf.186 | ✗138 | ✗138 | Platelets188, leukocytes158 |
Perf. Neuro‐Trace | Internal189 | ✗189 | Capillary186 | ✗189 | Other neuronal cells when fixed (brain)189 |
FITC | Passive42 | Perf.42 | ✗42 | ✗42 | None42 |
Tie2 | Surface190 | ✓64 | ✓* 64 | ✓* 64 | Monocytes, macrophages67, HSCs191 |
Dextran | Passive192 | Perf.192 | ✗192 | ✗192 | None193 |
VEGFR2, FLK‐1 | Surface194 | ✓194 | ✗139 | ✗139 | HSCs195, macrophages196, neurons197 |
VEGFR1, FLT‐1 | Surface198 | ✓198 | ✗199 | ✗199 | HSCs, macrophages196, neurons197 |
EC, endothelial cell, ECM, extracellular matrix; HSC, hematopoietic stem cell, MSC, mesenchymal stem cell, Perf., perfused; PC, pericyte, SMC, smooth muscle cell; Var., various. Labeling: yes (✓), no (✗).
Note: Table shows general trends, there are exceptions with specific tissues, species, and disease conditions. *See main text.
Figure 2.
Both endothelial cells and pericytes share markers used in the literature for labeling endothelial cells, and Col‐IV tracks, assumed to be regressed vessels, lack a pixel intensity profile indicative of a lumen. (A) Retina capillary with pericyte (NG2, red), IB4 lectin (green), endothelial cells (CD31, yellow), and cell nuclei (DAPI, cyan). (B) Retina capillary with pericyte and endothelial cells labeled with Col‐IV (green; scale bar 10 μm). (C) High‐resolution image of Col‐IV off‐vessel track (star) and lumenized blood vessel (arrow; scale bar 5 μm). (D) Comparison of Col‐IV relative pixel intensity profile across cross‐section of blood vessels and collagen tracks (P = 8.11E‐6, 2‐way analysis of variance, N = 10 vessels and tracks, error bars are standard deviation)
Another challenge for marking the microvasculature is identifying effective markers for PCs. Pericytes have no well‐established cross tissue exclusive marker, making them hard to target for analysis,58 although recent system‐level analyses have revealed novel candidate markers that await confirmation.59 A major point of contention with pericytes markers includes consensus with ASMA expression in pericytes, which is thought to be absent in pericytes on capillaries throughout some tissues, such as retina.60 However, recent evidence indicates that this may be a product of how tissue samples are processed: In the retina, it was shown that if standard fixation techniques are used with ASMA staining, capillary pericytes are ASMA‐negative,61 but if the samples were snap frozen with methanol fixation to prevent actin depolymerization, at least half of capillary pericytes are ASMA‐positive.61 We suspect that major portions of other canonically known ASMA‐negative pericyte populations across tissues might actually express this marker, and there is a possibility that ASMA is, in fact, a pan marker for pericytes that requires a more sensitive measurement technique to confirm. However, even if ASMA is expressed by all pericytes to some degree, it is not a unique marker for pericytes, because it is also expressed by vascular smooth muscle cells.
Finally, the expression of Tie2 by pericytes has been fiercely debated in the past decades, with extensive characterization of Tie2 expression in cultured pericytes,62 but a lack of Tie2 expression noted in pericytes in vivo.63 However, recent evidence has shown that a pericytes‐specific knockout of Tie2 leads to dramatically altered vascular structure, paired with a demonstration of Tie2 acting as a potent pericyte chemokine in vitro,64 together suggesting that Tie2 signaling may serve an important role in pericyte function. However, this finding awaits confirmation with the development of effective antibodies or other methods to directly label Tie2 in tissue and demonstrate pericyte expression in vivo. This controversy highlights the need to utilize measurement techniques that avoid such complications with variable results from tissue processing, such as fluorescent in‐situ hybridization65 which measures RNA expression of the target gene directly.
2.2. Animal models with endogenously labeled vasculature
An increasing number of transgenic murine models have been developed to visualize the microvasculature, including those that contain cell‐type specific fluorescent reporters for endothelial cells, smooth muscle cell, and pericytes (Table 2). We emphasize there is an important limitation of these reporter models that is often ignored: these animal models often only include the proximal endogenous reporter region with the fluorescent reporter, meaning that gene expression behavior from distal enhancers is often lost. An example of this is with Tie2 expression, which has been found in other cell types such as HSCs.66 and neutrophils, endothelial progenitor cells, macrophages,67 pericytes, 62 and keratinocytes.68 Yet the primary Tie2‐GFP mouse model is only known for GFP expression exclusively in endothelial cells,69 in this case serving as an advantage with a reporter line that appears to selectively label the vasculature and not track other cell types known to have endogenous expression.
Table 2.
Animal Models to Label Microvasculature
Name | Species | Sub Pop. | Cell Type Overlap |
---|---|---|---|
Tie2‐GFP | Ms, Zb200 | EC200 | None201 |
Tie2‐Cre‐GFP | Ms | EC202 | HSCs203 |
Tie2‐CreERT2‐EGFP | Ms | EC204 | None205 |
VE‐Cad‐Cre‐GFP | Ms | EC206 | Lymphatic endothelial cells206 |
VE‐Cad‐CreERT‐GFP | Ms | EC207 | None208 |
FLT1:GFP | Ms209, Zb210 | EC210 | Migratory angioblasts210 |
FLK1:GFP, RFP | Ms211, Zb212 | EC213 | None212 |
My11‐CreERT‐YFP | Ms | PC, SMC174 | None175 |
PDGFRB‐CreERT2‐TOM | Ms | PC,SMC214 | Glial cells214 |
NG2‐Dsred | Ms | PC, SMC156 | Oligodendrocyte progenitors215 |
NG2‐Cre‐ERT2M | Ms | PC, SMC216 | Oligodendrocyte precursors205 |
Tie1‐EGFP | Ms | Embryonic EC217 | None217 |
PDGFb‐CreERT2 | Ms | ECs218 | None218 |
β‐Actin‐GFP | Ms | EC, PC219 | Not examined205 |
α‐SMA‐GFP, RFP220 | Ms | Sub. PC, SMC221 | None221 |
α‐SMA‐CreERT2 | Ms | Sub. PC, SMC222 | None222 |
EC, endothelial cell; HSC, hematopoietic stem cell; PC, pericyte; SMC, smooth muscle cell. Species: Ms, mouse; Zb, zebrafish.
Note: Labeling can vary across tissues, and in most cases not rigorously verified.
2.3. In vitro and ex vivo models to study the microvasculature
Over the last century, various models have been used to study the complexity of the microvasculature, including those that utilize cultures of various cell populations, tissue explants, and animal models (Table 3). Historically, researchers have had to consider significant trade‐offs when choosing between different model systems. In vivo models typically have the best chance of recapitulating human disease since pathologies are heavily influenced by the complex interplay of a multitude of cell types.70 However, this benefit comes at a cost: In vivo models are usually limited by throughput, exhibit high cost in time and resources, and imaging techniques restricted by limitations with sedation duration, subject restraint, and detector scan speed. Furthermore, typically any intervention short of a cell‐specific knockout of an implicated gene will not establish cellular mechanism, which can take years to generate in an in vivo model. In vitro models typically exhibit much higher throughput and have a wider range of available analysis tools to characterize the system,71 but at the cost of greater simplification and abstraction of tissue structure and disease conditions compared to in vivo, such as lacking various cell types or blood flow.
Table 3.
Model Systems of Microvascular Architectures
Name/ Tissue | Type | Species | Throughput | Noninvasive Setup | Δ Vaso. Diameter | Timelapse | Adult | Angiogenesis | Network | Lumen | Flow | Mural |
---|---|---|---|---|---|---|---|---|---|---|---|---|
Chick Chorioallantoic Membrane | In vivo | Ch223 | ++ | ✗ | ✓ | ✓ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ |
Mesentery | In vivo | Ms224, Rt225, Ct226 | + | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Gluteus Maximus | In vivo | Ms227 | + | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Vessel Segments from Resistance Arteries | Ex vivo | Ms228, Hm229 | + | ✗ | ✓ | ✓ | ✓ | ✗ | ✓ | ✓ | ✗ | ✓ |
Skeletal Muscle | Ex vivo | Ms230, Rt231 | + | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Cremaster | In vivo | Ms232, Rt233 | + | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Retina | Ex vivo | Ms | + | ✓ | ✗ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Embryoid Explant | In vivo | Zb234, Ms235, Fg236 | ++ | ✗ | ✓ | ✓ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ |
Cornea Limbal | In vivo | Ms237 | + | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Microfluidic EC Chip | In vitro | Var238 | +++ | ✗ | ✗ | ✓ | ~ | ✓ | ✓ | ✓ | ✓ | ✗ |
EC‐PC Matrix Co‐culture | In vitro | Var239, 240 | ++++ | ✗ | ✗ | ✓ | ~ | ✗ | ✓ | ✓ | ✗ | ✓ |
EC‐PC Transwell | In vitro | Var.239 | ++++ | ✗ | ✗ | ✓ | ~ | ✗ | ✗ | ✗ | ✓ | ✓ |
EC Culture | In vitro | Var.239 | ++++ | ✗ | ✗ | ✓ | ~ | ✓ | ✓ | ✓ | ✓ | ✗ |
Dermal | In vivo | Ms44 | + | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ |
Developing Retina | In vivo | Ms.241 | + | ✗ | ✓ | ✗ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ |
Embryoid Body | In vivo | Ms.242 | +++ | ✗ | ✗ | ✓ | ✗ | ✓ | ✗ | ✗ | ✗ | ✓ |
Brain Explant | Ex vivo | Ms.243 | ++ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✗ | ✓ |
Retina Explant | Ex vivo | Ms.244, 245, Rt.246 | ++ | ✗ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✗ | ✓ |
Allantois Explant | Ex vivo | Ms.247 | ++ | ✗ | ✓ | ✓ | ✗ | ✓ | ✓ | ✓ | ✗ | ✓ |
EC Microbeads in Fibrin | In vitro | Var.248 | ++++ | ✗ | ✗ | ✓ | ~ | ✓ | ✓ | ✓ | ✗ | ✗ |
EC, endothelial cell; PC, pericyte. Species: Ch, chicken; Ct, cat; Fg, frog; Hm, hamster; Ms, mouse; Rt, rat; Var., various; Zb, zebrafish. Features: yes (✓), no (✗), various (~). Throughput: measure of degree of throughput for each protocol. Noninvasive setup: if model setup requires an invasive procedure. Δ Vaso. Diameter: if vasoconstriction or vasodilation can practically be examined in a real‐time fashion. Timelapse: if system can practically be imaged continuously in a real‐time fashion. Adult: if tissue analyzed is from adult or embryonic. Angiogenesis: if angiogenesis can be observed. Network: if model either has existing vascular network or can form a network. Lumen: whether vascular structures have a lumen. Flow: if vascular structures exhibit flow in model system. Mural: if smooth muscle cells and pericytes are included.
Note: Table is meant to capture general trends for what is feasible in a standard version of the experiment setup, there are exceptions.
However, the trade‐offs between in vivo and in vitro models are blurring now more than ever. Advances in new imaging techniques allow for in vivo imaging that provides the opportunity for higher throughput and fully temporal measurements in various tissues. The latest in vivo gene editing techniques, such as CRISPR/Cas9,72 have made targeted genetic alterations easier, yet there are still challenges remaining with regards to efficiency and off‐target binding of genetic payloads.73 At the same time, advances in 3D bioprinting, biomaterial research, and patient‐specific primary cell culture allow for more advanced in vitro models, although there is still difficulty with cell collection in these systems for subsequent analysis.71 Indeed, the number of available model systems has been growing, and with the advent of new analysis techniques, the opportunities to collect data from microvascular network architecture have increased dramatically and reveal new prospects for efficient and reproducible data capture.
3. STATE‐OF THE‐ART IMAGING MODALITIES FOR MICROVASCULAR NETWORKS
There are a vast range of techniques available for imaging the microvasculature, with trade‐offs between resolution, signal penetration, and acquiring multiplexed functional readouts, such as blood flow and tissue oxygenation (Table 4). Beyond the classical fluorescent‐based imaging modalities that have been a mainstay for imaging microvascular structure, there now exist several new technologies that also quantify microvascular function. Advances in photoacoustic microscopy have recently enabled imaging of a wide range of tissue depots, larger fields of view, and higher resolutions, along with capturing other functional data such as blood flow velocity and tissue oxygenation.74 The technology behind OCT, an imaging technique based on reflected light and measuring time of flight for photons, has recently improved with resolution to the point where these imaging modalities can successfully image the microvasculature.75 New super resolution imaging techniques developed in the last decade, such FPALM and stochastic optical reconstruction microscopy, have allowed visualization of structures in details beyond the resolution limit of visible light, allowing for direct imaging of individual proteins and flourophores.76
Table 4.
Imaging Modalities for Vascular Networks
Name | Resolution | Z Depth | Flow | Oxygen | Mechanism | Trade‐offs | |
---|---|---|---|---|---|---|---|
Z | XY | ||||||
Bright field | 1 μm249 | 0.25 μm249 | 50 μm38 | ✗ | ✗ | Absorbance 250 |
+ Visualize outlines of cells250, low cost + Temporal resolution − Excites fluorophores outside of imaging area250 − Image is blurred by emission from out‐of‐focus regions250 |
Fluorescent Widefield | 1 μm251 | 0.25 μm251 | 50 μm38 | ✗ | ✗ | Fluorescence 250 |
+ Temporal resolution: milliseconds252, low cost − High resolution requires immersion objectives251 − Image is blurred by emission from out‐of‐focus regions250 |
Point Scanning Confocal | 0.50 μm251 | 0.18 μm251 | 100 μm253 | ✗ | ✗ | Fluorescence 252 |
+ Pin‐hole reduce out‐of‐focus light250
+ Temporal resolution: milliseconds252 − Phototoxicity, photobleaching254 |
Spinning Disk Confocal | 0.75 μm254 | 0.25 nm254 | 100 μm38 | ✗ | ✗ | Fluorescence 252 |
+ Higher FPS compared to point scan250
+ Less phototoxic to cell, less photobleaching250 − Poorer filtering of out‐of‐focus light compared to point scan250 − Lower resolution, smaller FOV254 |
Two Photon | 0.4 μm251 | 0.20 μm251 | 1 mm255 | ✗ | ✗ | Fluorescent 256 |
+ Useful for thick specimens (>200um)250
+ Bleaching limited to imaging plane251, low light scatter256 + NIR light less phototoxic than VIS − Enhanced heating from NIR light − Broader excitation, pronounced photobleaching257 |
Photo‐acoustic | 1 μm258 | 10 μm258 | 5 mm259 | ✓ | ✓ | Flow, oxygenation 260 |
+ High contrast and spatial resolution, high framerate260
+ Imaging thick tissues (>1 cm)260 − Increased resolution at expense of ultrasonic penetration260 − Comparably low resolution260 |
Laser Doppler | 1 μm261 | 1 mm261 | ✓ | ✗ | Flow 262 |
+ Live imaging of flow262
− Long mapping time262 |
|
Laser Speckle | 10 μm263 | 1 mm264 | ✓ | ✗ | Fluorescence 265 |
+ Resolution adequate for low‐flow microvasculature265
+ Noninvasive, live imaging265, real‐time changes in flow265 − Requires knowledge of blood velocity distribution262 − Motion artifacts265 |
|
Second Harmonic | 2.5 μm266 | 0.70 μm266 | 300 μm267 | ✗ | ✗ | Auto‐fluorescence 268 |
+ Three‐dimensional resolution269, NIR wavelength + Label free267, Long imaging times269 − Low image quality in deep tissue270 |
Optical Coherence Tomography | 2 μm271 | 1 μm271, 272 | 2 mm253 | ✓ | ✗ | Reflectance 273 |
+ Temporal resolution: seconds252, label free274
+ Technology contained in endoscopes, handheld probes271 − Angiography visualizes only flow, not leakage274 |
RESOLFT, STED | 50 nm250 | 30 nm275 | 300 μm276 | ✗ | ✗ | Fluorescence 252 |
+ Video‐rate, live‐cell imaging252
+ Temporal resolution: seconds252 − Requires highly‐stable fluorophores252 |
FPALM | 50 nm250 | 20 nm275 | ✗ | ✗ | Fluorescence 252 |
+ Fast framerate for high resolution252, live‐cell imaging252
+ Image single molecules/single particle tracking252 − Requires photo‐switchable fluorophores252 |
|
Electron Microscopy | 8 nm277 | 1 nm252 | <1 μm278 | ✗ | ✗ | Fluorescence 252 |
+ High resolution252
− Limited labeling options19, no temporal resolution252, 279 − Restrictive sample prep.252, 279 |
Light‐Sheet | 0.75 μm280 | 0.25 μm280 | 10 mm264 | ✗ | ✗ | Fluorescence 280 |
+ Excellent optical sectioning 3D imaging281
+ Low bleaching and phototocicity281 − Restrictive sample prep281 |
4. MICROVASCULAR NETWORK ANALYSIS AND QUANTIFICATION
The microvascular network forms a sprawling architecture of interconnected vessels that vascularize nearly all tissues in the body. Such complex spatial networks undergo remodeling in adult tissue as well as embryonic; in quiescence as well as pathologic. Understanding changes in vessel morphology cannot be captured by a single metric to quantify its structure: A range of metrics must be used to summarize various unique characteristics of the network. While previous work has developed a multitude of metrics for quantifying microvascular architecture, we believe that further work must be done in both developing new metrics and demonstrating that a given set of available metrics provide unique and useful information to answer biological questions. To this end, a series of suggestions are proposed to increase scientific rigor and reproducibility of quantifying the complexities of the microvasculature.
4.1. Metrics for quantifying microvascular networks
Previous research has developed various metrics for microvascular network analysis (Figure 3A‐G), including the fraction of image area composed of blood vessels (VAF),77 blood vessel length normalized by image field of view (vessel length density),78 average vessel diameter of all vessels or divided by vessel type,79 density of branchpoints,80 tortuosity,81 lacranuity,78 fractal dimension,81 and max extra‐vascular diffusion distance to examine tissue oxygen perfusion.82 Other metrics have been developed outside of this set but not standardized and adopted by consensus. Studies often normalize metrics in different ways, such as measuring branchpoints per image, normalizing to field of view, or normalizing to vessel length. We posit that metrics should be designed to encourage valid comparisons across research studies and should be normalized to facilitate this process. Thus, using a simple metric of vessel length83 is not as useful as vessel length density, a metric that can be directly compared over a range of spatial resolutions and imaging modalities.
Figure 3.
Basic metrics quantifying the complexities of the microvascular architecture. Visual explanation of metrics that have been used to quantify various aspects of microvessel network architecture, including (A) VAF, (B) vessel length density, (C) vessel diameter, (D) branchpoints density, (E) tortuosity, (F) lacranuity and fractal dimension, (G) extra‐vascular diffusion distance, and (H) vessel segment partitioning
4.2. Architectural features of microvasculature: bridging form and function
Capillary architecture possesses markedly different structures to meet the unique metabolic demands of peripheral tissues,84 including the radial spoke‐wheel structure of the retina, parallel beds of skeletal muscle, or dense networks of the liver (Figure 4A‐F). Even within a single tissue such as the retina, there is impressive heterogeneity in microvascular architecture between tissue locations (Figure 4G‐I). This tissue environment heterogeneity is further reflected by unique endothelial transcriptomes found in each organ85 and distinct endothelial marker profiles at different parts of the vascular tree.86
Figure 4.
Heterogeneity of blood vessel network structure across and within tissue. IB4 lectin Perfused microvessels of (A) heart, (B) diaphragm, (C) skeletal muscle, (D) liver, (E) peritoneal cavity, (F) inguinal fat, and of the three distinct vascular layers of the retina (G‐I; scale bar 50 μm)
With much of biology, function and form are closely interwined87: the microvasculature is no exception. A wide range of biological behaviors, including blood vessel growth, regression, dilation, constriction, stability, and permeability, can be mapped to quantitative metrics of microvascular structure and give insight into physiologic and pathological function of the microcirculation (Figure 5). Beyond the adaptations of microvessel networks to support unique tissue metabolic environments, morphological changes in vessel structure are hallmarks of key vascular remodeling events. Spatial distribution of capillary networks determines spatial heterogeneity of oxygenation and nutrient delivery.88, 89 Enriched build‐up of extra‐cellular matrix can indicate a fibrotic response90 to inflammatory conditions. Increased blood vessel tortuosity can indicate signs of endothelial cell activation and pathological microvascular remodeling and/or ischemia‐induced arterialization in collateral microvessels.91
Figure 5.
Bridging form and function: correspondence between microvasculature architecture metrics and biological behaviors. Schematized microvasculature network with various cellular and acellular components (multicolored font) mapped to quantitative image analysis metrics that indicate different aspects of microvascular function (black font)
Moreover, certain quantification metrics may carry unique significance depending on the location analyzed within the vascular tree. For instance, changes in vessel diameter in arterioles suggest changes in vascular smooth muscle cell vasoconstriction to modulate vascular resistance,92 capillary diameter changes are indicative of changes to pericyte contraction and distribution of flow and oxygenation,93 and venule dilation suggests changes to blood capacitance (storage)94 or remodeling in response to inflammation to facilitate leukocyte extravasation from circulation.95
4.3. The need for pairing perivascular and microvascular analysis
Analyzing changes to the perivascular space can yield just as important insights as the vasculature itself given the close cross‐talk between endothelial cells and smooth muscle cell and pericytes. For example, changes in pericyte density are known to play a key role in the pathogenesis of diabetes,27 and changes in pericyte locations relative to branchpoints96 have been associated with changes to stability and sensitization of the microvascular network. We emphasize the need to analyze perivascular behavior as well as microvascular remodeling to truly understand the structure and function of microvascular networks: A common limitation of many studies is to study one or the other in isolation. Most existing software for quantifying microvascular network architecture accomplishes this by evaluating the endothelial network structure. There is one software package that can analyze perivascular cell recruitment to the vasculature through overlap of the two structures.97 While this metric may be confounded by changes in perivascular or vascular density across study groups, and perivascular cells may associate with the vasculature with minimal channel overlap, this software allows researchers to begin to probe perivascular interactions with the microvasculature. A survey of the published literature reveals that the most common approach for analyzing perivascular cell coverage and/or morphology (eg, of pericytes,98 smooth muscle cell,99 or other cell types like macrophages100 that are known to play key roles in angiogenesis and vessel remodeling101) is through manual or basic automated comparisons of thresholded area or nuecleii.102 For example, basic cell counts can be obtained manually through ImageJ's cell103 counting feature, the multipoint tool,104 or using its particle analysis set of tools for automated analysis.104 More extensive positional or morphological investigations of perivascular structures require custom image analysis solutions that have yet to be developed. Automated and quantitative analysis of microvascular networks paired with detailed analysis of smooth muscle and pericyte cell populations could become a standard pipeline that would enable better understanding of microvascular remodeling mechanisms and the development of new therapeutics for microvascular diseases.
4.4. Software packages for quantifying microvascular networks
Alterations in microvessel network architecture have been used ubiquitously in studying vascular diseases, and there are a multitude of software packages available for quantifying changes in architecture (Table 5). Three, notably, have been used in a significant number of publications, namely AngioQuant, AngioTool, and RAVE.
Table 5.
Vessel Network Analysis Tools
Name | Lang. | Application | Metrics | Validation | Cts | Yr | Cts/yr |
---|---|---|---|---|---|---|---|
AngioQuant83 | MATLAB | Brightfield, Cell Culture | Segment Count, VL, VA, VAF, Segment Area, BP, BP/Segment Count | QT BD | 104 | 2005 | 8 |
RAVE81 | MATLAB | Fluorescent, Tissue | VAF, VLD, VR, FD | QT manually analyzed BD for VAF, VLD, and VR. Basic In silico for FD | 24 | 2011 | 3.4 |
Vessel Analysis ImageJ Macro | ImageJ Macro | Brightfield, Tissue | VAF, VLD | None | ‐ | ‐ | ‐ |
AngioTool78 | Java, ImageJ | Fluorescent, Tissue | VAF, SC, VL, VLD, EP, lacunarity, BP/area | QL manually analyzed BD | 111 | 2011 | 15.9 |
CSAQ282 | Web Based | Brightfield, Tissue | VL, VAF, BP; Contrast, Entropy | Manual (BP), QT comparison to AngioQuant | 24 | 2008 | 2.4 |
Breast Tumor Microvasculature Reconstruction82 | MATLAB, JAVA | Micro Ct, Tumor | VLD, VD, SL, MEVDD, FD, Tortuosity, Flow | Metrics from BD within literature values | 17 | 2013 | 3.4 |
VESGEN 2D283 | Java, ImageJ | Brightfield/ Flourescence, Tissue | VD, VT, FD, VAF, VLD, BP | QT BD | 29 | 2009 | 3.2 |
BD, biological data; BP, branchpoints; EP, endpoints; FD, fractal dimension; MEVDD, max extra‐vascular diffusion distance; NS, not specified; QL, qualitative; QT, quantitative; SC, segment count; SL, segment length; VLD, vessel length density; VD, vessel diameter; VR, vessel radius.
AngioQuant has been developed to analyze endothelial networks in vitro, with a focus on quantifying various metrics of tubule formation using bright field images.83 Recently, it has been adapted for use in evaluating higher resolution datasets of microvascular networks in vivo105 and in histological samples.106 Its validation is focused on quantification of in vitro experiments showing trends of changes with various metrics, but no statistical comparisons between study groups. The datasets were not validated against manually analyzed images and there is no analysis included comparing accuracy and overall performance between AngioQuant and other available software packages.
AngioTool is presented as a quick, hands‐off, and reproducible image analysis tool, deployed as an ImageJ plugin, for quantification of microvascular networks in microscopic images.78 The validation of AngioTool included analyzing biological data from murine hindbrain and retina using various metrics, including visualized vessel segmentation, vessel centerline, and branchpoints presented for qualitative inspection. For quantitative biological validation, endothelial cell explants were cultured and analyzed with two drug treatments that were known to alter vascular structure as a positive control.78 Additionally, output metrics were validated with a subset of manually counted images in an unblinded fashion with two investigators.
RAVE is an image analysis tool that can be used on a wide array of images81 to accelerate the unbiased, quantitative analysis of microvasculature architecture. Validation for this tool included comparing automated outputs generated by RAVE to manual analysis of various morphometric parameters for in vivo microvessel networks in murine spinotrapezius muscle tissue and in a xenograph tumor model. A dataset of images was compared to manually processed images with a Bland‐Altman analysis.
4.5. Improving vessel quantification, analysis, and interpretation
We estimate that these software packages are largely underutilized, based on the high number of published manuscripts that refer to the quantification of microvessel architecture. Indeed, a search on PubMed on relevant terminology (terms used included: microvasculature density, capillary dropout, pericyte dropout; see all terms in Appendix S1) reveals over 120 000 publications to date. While this query includes publications that merely mention the terms searched for, the nearly three order‐of‐magnitude difference between citations of these software programs compared to this large collection of publications suggests that there is an unmet need for vessel architecture analysis beyond the available options, with researchers often resorting to manual ad hoc analysis of microvessel networks, leading to decreased repeatability, comparability, and scientific rigor. We propose the following design criteria for an effective software package:
Ground truth validation: A rigorous and complete validation of software requires a comprehensive analysis of multiple types of datasets. This includes an extensive comparison of automated results to manually processed images, not just with output metrics, but also with the pixel‐by‐pixel raw segmentation, skeleton centerline, and branchpoints locations quantified with a combination of false positive rates, false negative rates, Bland‐Altman analysis, and SSR. Ideally, this manual comparison would include multiple study groups with known vascular differences in architecture between them.
Biological validation: Validating the automated pipeline with several biological datasets with known differences (biological positive controls), ideally from different tissue and/or imaging resolutions, will demonstrate the efficacy of the program in practice. This analysis demonstrates that the program can detect true positives in actual dataset, where a real change is detected between study groups.
In silico validation: Program development needs to be paired with a validated parameterized computational model that can generate artificial in silico vascular networks107 to verify that changing basic parameters of the model yields expected changes in metric output with the image analysis pipeline.
Quantitative comparisons between previously developed programs: The field benefits far less from releasing another “one‐off” vessel image analysis program independent from previous work and instead should test and demonstrate its efficacy compared to existing software. Outputs from each program and manual analysis should be compared, including output metrics, raw segmentation, skeletonization, and branchpoints assignment in the form of false positive and negative error rates, Bland‐Altman analysis, and SSR. Execution time should also be critically evaluated and reported, given the importance of balancing throughput with accuracy.
Standardized metric sets: Each software program has a unique collection of metrics that are often calculated using different methods. A consensus of metrics needs to be established in the interest of rigor and reproducible science. An example of this would be measuring branchpoints: Some packages display raw counts, while others normalize to image area or vessel length.
Effectiveness of metric collections: Methods should be developed to not only test and validate each method but also demonstrate their usefulness as a collection in determining changes in the vascular architecture. Analysis needs to be done to show that these new metrics provide unique non‐correlative information compared to existing metrics, utilizing techniques developed from the field of feature selection.108
Effect of image quality on output metrics: An examination needs to be performed with respect to how the program performs when image quality varies between study groups from batch effects, or simply has a high degree of variance. Image quality of the datasets provided with these software packages often appears ideal. High variance with image quality may skew segmentation results and output metrics, so output metrics across a range of image qualities should be examined.
Effect of parameter adjustment: Some software packages allow for adjustment of key image processing parameters to enhance results, but the effect (and bias) of allowing the user to freely alter image processing outcomes needs to be rigorously examined and reported.
Blinded image analysis: Software packages should include built‐in support for image filename anonymization to blind the user from an image's study group assignment to minimize bias as images are analyzed.
Semi‐automated curation: Image quality and marker expression can change between study groups, which could bias automated segmentation and results. Image analysis programs should, therefore, build on previous efforts97 to include the option to efficiently curate segmentation within a study group in a blinded fashion in regions where automated analysis fails. While some of the programs allows for a degree of curation with adjusting image processing parameters, there is no examination with how this can bias results if the researcher changes these parameters between study groups or images.
Built‐in detection of insufficient sampling: It is important for the image datasets to sample enough of the microvasculature for valid metric quantification. For simple metrics, such as vessel length density or VAF, a simple examination of variance and power analysis can determine whether more images of a biological sample are required. However, for more advanced quantification metrics, such as lacunarity and fractal dimension, or graph theory‐based metrics, the metrics become nonsensical if the field of view is too small and not enough vascular network sampled. These metrics need to be studied and predictive algorithms developed to warn the user if the dataset is at risk for yielding invalid results for advance structural metrics.
Source code and dataset availability: While most published software packages will provide source code and data upon request, we believe that it should be standard to make both freely available for download on long‐term hosting platforms such as Github, Bitbucket, or an institutional repository. This removes any barrier to iterate on previous work and facilitates comparison of software packages. Freely available image datasets will also standardize the validation process and enhance progress within the field in the same way standard datasets have with analyzing retina microvessels in fundus imaging.109, 110
Novel metrics: More metrics need to be developed to describe all observed complexities found in the network structure, with the long‐term view that a sufficient number of metrics for characterizing the microvasculature would allow for the successful creation of in silico artificial networks that are indiscernible from experimentally derived microvascular network structures. Any degree short of this reproduction would mean that information is being lost by the current metric set. Techniques for developing new metrics can be guided from the field of feature engineering.111
Without tackling these issues, new software programs are merely presented to research scientists “as is” without allowing them to make informed decisions on how to produce high‐quality unbiased results. We argue that until these issues are dealt with, the use of these packages has the risk of leading to a significant error rate in microvascular research: where the software reveals a positive error with a quantifiable change between groups where none existed, or even worse, where research is not pursued based on a negative error where no change is observed between study groups where one exists.
4.6. Applications of machine learning, graph theory, and modeling in quantifying microvascular architecture
Metric effectiveness not only needs to be evaluated on an individual basis, but the effectiveness of a given collection of metrics in combination needs to be evaluated. This can start with examining covariance matrices of metrics from in silico and biological datasets to evaluate how much unique information they bring relative to one another. Effectiveness of metric sets could be evaluated using principal components analysis, partial least squares, or more advanced methods of feature selection techniques from the field of feature engineering,108 especially when applied to in silico artificial networks where basic parameters (vessel length density, vessel diameter, branchpoints density, tortuosity, connectivity) can be changed in a controlled fashion. Further research must be conducted on what makes an effective metric in an unbiased fashion, and consensus much be reached on normalization of metrics so they can be used in a standardized way, such as the disagreement on how to normalize branchpoints counts (by image area, vessel length, or binned vessel diameter). This will depend on the development and pairing of fully parametrized in silico models112 (Table 6), structural models of models of vascular flow,113, 114 and models of predictive tissue oxygenation115 with biological experiments116 to fully connect metrics of microvascular structure to biological behaviors.
Table 6.
Computation Models of Microvascular Architecture
Description | Language | Method | Type | Validation |
---|---|---|---|---|
Particle‐based EC Network284 | C++ | ABM, CPM | Vasculogenesis | QT to classic CPM |
Retina ABM285 | NetLogo | ABM | Retinal developing vasculature | QT comparison retinal BD |
3D CPM of Tumor Growth286 | CompuCell3D | ABM, CPM | Tumor angiogenesis | QL to Macklin et al.287 |
ABM for Disruption of Vasculogenesis288 | CompuCell3D | ABM | Vasculogenesis | QT to BD with AngioTool78 |
3D Sprouting Angiogensis289 | NS | ABM | Sprouting angiogenesis | QT metrics within BD range |
Hypoxic Vessel Sprouting290 | NS | ABM Hybrid | Any 2D or 3D vessel formation in a tissue | QT comparison with BD |
Tumor Angiogenesis and Patterning291 | NS | ABM Hybrid | Sprouting angiogenesis | QT metrics within BD range |
Angiogenesis with Discrete Random Walks292 | NS | ABM Hybrid | Tumor angiogenesis | QL assessment of simulation results |
ABM of Tumor Angiogenesis and Regression293 | NS | ABM Hybrid | Tumor angiogenesis and regression | QL to BD |
3D Phase Field ABM of Vascular Networks294 | NS | ABM Hybrid | 3D Angiogenesis | QL to basic theoretical behavior |
Adaptive Network with Flow295 | C | Stochastic Hybrid model | Flow, oxygen transport, and adaptation of existing network | Thorough quantitative comparison to experimental results across all categories |
Multiphase Tumor Angiogenesis Growth296 | CAST3M | Continuum Discrete | Tumor growth | QT comparison with BD |
Vessel Generator for Cell‐colocalization (CIRCOAST)135 | MATLAB | Structural Descriptive Model | Static adult microvascular network for basic model validation | QT metrics within BD range |
Tumor Angiogenesis with Blood and Interstitial Flow297 | NS | Hybrid | Tumor growth | QT metrics within BD range |
ABM, Agent‐based model; BD, biological data; CPM, cellular pots model; QL, qualitative; QT, quantitative; NS, not specified.
Blood vessel networks can be abstracted as a series of branchpoints, or nodes, with varying connectivity with each other. Previous work has explored the basic concept of abstracting the microvasculature as a graph,117 and we believe there are a wealth of relevant metrics that could be applied from quantifying graph networks (Figure 6). Examples include metrics measuring centrality of each node,118 scoring the relative importance of edges in connecting nodes, connective redundancy,119 and information diffusion.120 Machine learning methods such as convolution neural networks and other techniques121 have also been applied to graph networks.122 Previous applications of network tomography outside of biomedical research,123 such as internet tomography,124 may be a pertinent source of applicable methods for characterizing complex microvascular networks.
Figure 6.
Novel image analysis metrics by analyzing the microvasculature with graph theory. A, Confocal microscopy image of the murine retinal deep vasculature, with CD105 marking ECs (white; scale bar = 50 μm). B, Image analyzed with basic segmentation, skeletonization, and branchpoints classification, with vessel centerline/skeleton (white), branchpoints (red), and EP (turquoise). C, Conversion of vasculature into a graph, with branchpoints indicated as “nodes” (blue) and vessel segments as “edges” (gray). D, Visual summary of types of metrics to quantify graphs, with removed edges representing change to network after a blood vessel has regressed or experiences obstructed flow
4.7. Relevant techniques from related network architectures
Analysis of full feature microvascular architectures can also benefit from adapting techniques used to analyze similar network structures. A prime example is fundus imaging of human retinal vascular networks: Although such images fail to resolve capillary portions of the microvascular network, this application has been extensively explored due to its established clinical significance in evaluating eye disease and a collection of systemic diseases,125 with a plethora of methods developed to segment, quantify, and validate vessel architecture.126 Indeed, a machine learning‐based classification pipeline for fundus images has recently been approved by the FDA for diagnosis of diabetic retinopathy.127 Methods used in the processing and quantification of neuronal network image datasets128 may yield useful techniques for analyzing microvascular networks.129 In vivo clinical imaging techniques, such as micro‐CT130 of lung vascular networks, may also yield insight into extending 2D imaging and quantification into the third dimension.131
Although this review focuses on 2D image quantification techniques, many of the imaging modalities mentioned acquire data in 3D directly, or through a series of 2D slices. Over the long term, the field would most benefit from acquiring and analyzing 3D datasets to eliminate any confounding phenomena that arises from analyzing a 2D‐projected representation of complex 3D structures. Such 2D abstractions can lead to altered metrics, such as false branch points where vessels appear to overlap in 2D but exist at distinct elevations in 3D, introducing error to other metrics such as segment length. Furthermore, the 3D orientation of vessel segments relative to one another is especially important when characterizing local tissue oxygenation.89 An in‐depth examination of how 2D structural metrics can characterize a projected 3D structure like a microvascular network would also be necessary to understand the trade‐offs and reveal what information is missed with this simplification.
4.8. Statistics to analyze microvascular interactions: beyond generic
The metrics covered in this review require basic 2‐sample or multi‐sample statistical tests to determine whether there is a difference in structure and morphology of vascular networks between study groups. Yet there are also new statistics being developed based on modeling null distributions (output if the null hypothesis is true and there is no difference between groups) that could be extended to quantifying the microvascular architecture. A prime example of this is a technique to measure cellular recruitment with a given cell type and the vascular network,132 that maintains validity in conditions where generic statistics fail. While cell recruitment has been analyzed,97 previous metrics of cell‐to‐cell colocalization events fails to properly measure changes in cell recruitment if there are changes to vascular density or cell density across study groups. This confounding phenomenon will lead to false positives when testing between study groups132: instances where the test indicates there is a significant change in cell recruitment, when in reality there is none. Null modeling of random cell placement is used to avoid the deficiencies of generic statistics, and we believe this modeling approach could be applied to evaluating perivascular cell recruitment to blood vessel architecture using an in silico model and provide researchers with a more robust statistical hypothesis test for analyzing microvessel architecture.
PERSPECTIVES
The microvasculature is implicated in pathogenesis and maintenance of the deadliest maladies of the modern world. Understanding microvasculature's function, adaptation, and contribution to disease is enabled by the application of metrics that quantify changes to microvascular network architecture in in vivo, in vitro, and in silico model systems. We highlight opportunities to further the field by improving scientific rigor and reproducibility through the development and validation of software that reliably, comprehensively, and in an automated manner, characterizes the complexities of microvascular architecture using pre‐existing and novel metrics.
Supporting information
Corliss BA, Mathews C, Doty R, Rohde G, Peirce SM. Methods to label, image, and analyze the complex structural architectures of microvascular networks. Microcirculation. 2019;26:e12520 10.1111/micc.12520
Funding information
NIH‐U01AR069393, NIH‐U01HL127654, The Hartwell Foundation, The Stanford Allen Discovery Center (to S.M.P.), and NIH GM090033 (to G.R.).
REFERENCES
- 1. Jacob M, Chappell D, Becker BF. Regulation of blood flow and volume exchange across the microcirculation. Crit Care. 2016;20:319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immunity. 2014;41(5):694‐707. [DOI] [PubMed] [Google Scholar]
- 3. Tonnesen MG, Feng X, Clark RAF. Angiogenesis in wound healing. J Investig Dermatol Symp Proc. 2000;5(1):40‐46. [DOI] [PubMed] [Google Scholar]
- 4. Chambers R, Zweifach BW. Intercellular cement and capillary permeability. Physiol Rev. 1947;27(3):436‐463. [DOI] [PubMed] [Google Scholar]
- 5. Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010;9(7):689‐701. [DOI] [PubMed] [Google Scholar]
- 6. Camici PG, Crea F. Coronary microvascular dysfunction. N Engl J Med. 2007;356(8):830‐840. [DOI] [PubMed] [Google Scholar]
- 7. Tooke JE. Microvascular function in human diabetes. A physiological perspective. Diabetes. 1995;44(7):721‐726. [DOI] [PubMed] [Google Scholar]
- 8. Centers for Disease Control and Prevention . Deaths: Final Data for 2015. National Vital Statistics Reports. 2017; 66(6):75. [PubMed] [Google Scholar]
- 9. Zhang H, van Olden C, Sweeney D, Martin‐Rendon E. Blood vessel repair and regeneration in the ischaemic heart. Open Heart. 2014;1(1):e000016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Ostergaard L, Kristiansen SB, Angleys H, et al. The role of capillary transit time heterogeneity in myocardial oxygenation and ischemic heart disease. Basic Res Cardiol. 2014;109(3):409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Rosenkranz S, Gibbs JSR, Wachter R, De Marco T, Vonk‐Noordegraaf A, Vachiéry J‐L. Left ventricular heart failure and pulmonary hypertension. Eur Heart J. 2016;37(12):942‐954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. García‐Román J, Zentella‐Dehesa A. Vascular permeability changes involved in tumor metastasis. Cancer Lett. 2013;335(2):259‐269. [DOI] [PubMed] [Google Scholar]
- 14. Au SH, Storey BD, Moore JC, et al. Clusters of circulating tumor cells traverse capillary‐sized vessels. Proc Natl Acad Sci USA. 2016;113(18):4947‐4952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Voelkel NF, Cool CD. Pulmonary vascular involvement in chronic obstructive pulmonary disease. Eur Respir J. 2003;22(46 suppl):28s‐32s. [DOI] [PubMed] [Google Scholar]
- 16. Eliason G, Abdel‐Halim SM, Piehl‐Aulin K, Kadi F. Alterations in the muscle‐to‐capillary interface in patients with different degrees of chronic obstructive pulmonary disease. Respir Res. 2010;11(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Widdicombe JG. Asthma. Tracheobronchial vasculature. Br Med Bull. 1992;48(1):108‐119. [DOI] [PubMed] [Google Scholar]
- 18. Østergaard L, Jespersen SN, Mouridsen K, et al. The role of the cerebral capillaries in acute ischemic stroke: the extended penumbra model. J Cereb Blood Flow Metab. 2013;33(5):635‐648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Østergaard L, Jespersen SN, Engedahl T, et al. Capillary dysfunction: its detection and causative role in dementias and stroke. Curr Neurol Neurosci Rep. 2015;15(6):37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Yemisci M, Gursoy‐Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative‐nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15(9):1031‐1037. [DOI] [PubMed] [Google Scholar]
- 21. Gjedde A, Kuwabara H, Hakim AM. Reduction of functional capillary density in human brain after stroke. J Cereb Blood Flow Metab. 1990;10(3):317‐326. [DOI] [PubMed] [Google Scholar]
- 22. Adair TH, Montani J‐P. Overview of Angiogenesis. Morgan & Claypool Life Sciences; 2010. https://www.ncbi.nlm.nih.gov/books/NBK53238/. Accessed July 30, 2018. [PubMed] [Google Scholar]
- 23. Fogelson AL, Neeves KB. Fluid mechanics of blood clot formation. Annu Rev Fluid Mech. 2015;47:377‐403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Muller WA. Getting leukocytes to the site of inflammation. Vet Pathol. 2013;50(1):7‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Østergaard L, Nagenthiraja K, Gyldensted L, et al. Capillary dysfunction in Alzheimer's disease. Alzheimers Dement. 2013;9(4):P269‐P270. [Google Scholar]
- 26. Hecht M, Krämer LM, von Arnim CAF, Otto M, Thal DR. Capillary cerebral amyloid angiopathy in Alzheimer's disease: association with allocortical/hippocampal microinfarcts and cognitive decline. Acta Neuropathol. 2018;135(5):681‐694. [DOI] [PubMed] [Google Scholar]
- 27. Beltramo E, Porta M. Pericyte loss in diabetic retinopathy: mechanisms and consequences. Curr Med Chem. 2013;20(26):3218‐3225. [DOI] [PubMed] [Google Scholar]
- 28. Moldoveanu B, Otmishi P, Jani P, et al. Inflammatory mechanisms in the lung. J Inflamm Res. 2008;2:1‐11. [PMC free article] [PubMed] [Google Scholar]
- 29. Gurka DP, Balk RA. Acute respiratory failure In: Parrillo JE, Dellinger RP, eds. Critical Care Medicine, 3rd edn Philadelphia, PA: Mosby; 2008:773‐794. http://www.sciencedirect.com/science/article/pii/B9780323048415500406. Accessed July 30, 2018. [Google Scholar]
- 30. Azzi S, Hebda JK, Gavard J. Vascular permeability and drug delivery in cancers. Front Oncol. 2013;3:211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Simone E, Ding B‐S, Muzykantov V. Targeted delivery of therapeutics to endothelium. Cell Tissue Res. 2009;335(1):283‐300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Germain RN. Maintaining system homeostasis: the third law of Newtonian immunology. Nat Immunol. 2012;13:902‐906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Opferman JT, Korsmeyer SJ. Apoptosis in the development and maintenance of the immune system. Nat Immunol. 2003;4(5):410‐415. [DOI] [PubMed] [Google Scholar]
- 34. Parkin J, Cohen B. An overview of the immune system. Lancet. 2001;357(9270):1777‐1789. [DOI] [PubMed] [Google Scholar]
- 35. Gramatikov BI. Modern technologies for retinal scanning and imaging: an introduction for the biomedical engineer. BioMed Eng OnLine. 2014;13:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Fabrikant SI, Maggi S, Montello DR. 3D network spatialization: does it add depth to 2D representations of semantic proximity? In: Duckham M, Pebesma E, Stewart K, Frank AU, eds. Geographic Information Science. Basel: Springer International Publishing; 2014:34‐47. (Lecture Notes in Computer Science). [Google Scholar]
- 37. Miolane N, Pennec X. A survey of mathematical structures for extending 2D neurogeometry to 3D image processing; 2015. https://hal.inria.fr/hal-01203518/document. Accessed July 30, 2018.
- 38. Combs CA. Fluorescence microscopy: a concise guide to current imaging methods. Curr Protoc Neurosci. 2010;Chapter 2:Unit2.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Nuzzo R. Scientific method: statistical errors. Nature. 2014;506(7487):150. [DOI] [PubMed] [Google Scholar]
- 40. Bergijk EC, Van Alderwegen IE, Baelde HJ, et al. Differential expression of collagen IV isoforms in experimental glomerulosclerosis. J Pathol. 1998;184(3):307‐315. [DOI] [PubMed] [Google Scholar]
- 41. Daly SM, Leahy MJ. ‘Go with the flow ‘: a review of methods and advancements in blood flow imaging. J Biophotonics. 2013;6(3):217‐255. [DOI] [PubMed] [Google Scholar]
- 42. Hoffmann A, Bredno J, Wendland M, Derugin N, Ohara P, Wintermark M. High and low molecular weight fluorescein isothiocyanate (FITC)–dextrans to assess blood‐brain barrier disruption: technical considerations. Transl Stroke Res. 2011;2(1):106‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Robertson RT, Levine ST, Haynes SM, et al. Use of labeled tomato lectin for imaging vasculature structures. Histochem Cell Biol. 2015;143(2):225‐234. [DOI] [PubMed] [Google Scholar]
- 44. Gurevich DB, Severn CE, Twomey C, et al. Live imaging of wound angiogenesis reveals macrophage orchestrated vessel sprouting and regression. EMBO J. 2018;4:e97786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Siegenthaler JA, Choe Y, Patterson KP, et al. Foxc1 is required by pericytes during fetal brain angiogenesis. Biol Open. 2013;2(7):647‐659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Boscia F, Esposito CL, Casamassa A, de Franciscis V, Annunziato L, Cerchia L. The isolectin IB4 binds RET receptor tyrosine kinase in microglia. J Neurochem. 2013;126(4):428‐436. [DOI] [PubMed] [Google Scholar]
- 47. Allinson KR, Lee HS, Fruttiger M, McCarty J, Arthur HM. Endothelial expression of TGFβ type II receptor is required to maintain vascular integrity during postnatal development of the central nervous system Chen J, editor. PLoS ONE. 2012;7(6):e39336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Hu J, Popp R, Frömel T, et al. Müller glia cells regulate Notch signaling and retinal angiogenesis via the generation of 19,20‐dihydroxydocosapentaenoic acid. J Exp Med. 2014;211(2):281‐295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Dominguez E, Raoul W, Calippe B, et al. Experimental branch retinal vein occlusion induces upstream pericyte loss and vascular destabilization. PLoS ONE. 2015;10(7):e0132644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Fan J, Ponferrada VG, Sato T, et al. Crim1 maintains retinal vascular stability during development by regulating endothelial cell Vegfa autocrine signaling. Development (Cambridge, England). 2014;141(2):448‐459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Lam S, van der Geest RN, Verhagen NAM, Daha MR, van Kooten C. Secretion of collagen type IV by human renal fibroblasts is increased by high glucose via a TGF‐beta‐independent pathway. Nephrol Dial Transplant. 2004;19(7):1694‐1701. [DOI] [PubMed] [Google Scholar]
- 52. Baluk P, Morikawa S, Haskell A, Mancuso M, McDonald DM. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol. 2003;163(5):1801‐1815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Brown WR. A review of string vessels or collapsed, empty basement membrane tubes. J Alzheimers Dis. 2010;21(3):725‐739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. McVicar CM, Ward M, Colhoun LM, et al. Role of the receptor for advanced glycation endproducts (RAGE) in retinal vasodegenerative pathology during diabetes in mice. Diabetologia. 2015;58(5):1129‐1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Yang P, Pavlovic D, Waldvogel H, et al. String vessel formation is increased in the brain of parkinson disease. J Parkinsons Dis. 2015;5(4):821‐836. [DOI] [PubMed] [Google Scholar]
- 56. Abramsson A, Lindblom P, Betsholtz C. Endothelial and nonendothelial sources of PDGF‐B regulate pericyte recruitment and influence vascular pattern formation in tumors. J Clin Investig. 2003;112(8):1142‐1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Winkler EA, Sengillo JD, Bell RD, Wang J, Zlokovic BV. Blood–spinal cord barrier pericyte reductions contribute to increased capillary permeability. J Cereb Blood Flow Metab. 2012;32(10):1841‐1852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kelly‐Goss MR, Sweat RS, Stapor PC, Peirce SM, Murfee WL. Targeting perciytes for angiogenic therapies. Microcirculation (New York, N.Y. : 1994). 2014;21(4):345‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. He L, Vanlandewijck M, Raschperger E, et al. Analysis of the brain mural cell transcriptome. Sci Rep. 2016;6:35108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Hughes S, Chan‐Ling T. Characterization of smooth muscle cell and pericyte differentiation in the rat retina in vivo. Invest Ophthalmol Vis Sci. 2004;45(8):2795‐2806. [DOI] [PubMed] [Google Scholar]
- 61. Alarcon‐Martinez L, Yilmaz‐Ozcan S, Yemisci M, et al. Capillary pericytes express α‐smooth muscle actin, which requires prevention of filamentous‐actin depolymerization for detection. eLife. 2017;7:1‐17. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5862523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Cai J, Kehoe O, Smith GM, Hykin P, Boulton ME. The angiopoietin/Tie‐2 system regulates pericyte survival and recruitment in diabetic retinopathy. Invest Opthalmol Vis Sci. 2008;49(5):2163. [DOI] [PubMed] [Google Scholar]
- 63. Park DY, Lee J, Kim J, et al. Plastic roles of pericytes in the blood–retinal barrier. Nat Commun. 2017;8:15296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Teichert M, Milde L, Holm A, et al. Pericyte‐expressed Tie2 controls angiogenesis and vessel maturation. Nat Commun. 2017;8:16106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Fe Lanfranco M, Loane DJ, Mocchetti I, Burns MP, Villapol S. Combination of fluorescent in situ hybridization (FISH) and immunofluorescence imaging for detection of cytokine expression in microglia/macrophage cells. Bio Protoc. 2017;7(22):1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Arai F, Hirao A, Ohmura M, et al. Tie2/angiopoietin‐1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149‐161. [DOI] [PubMed] [Google Scholar]
- 67. Patel AS, Smith A, Nucera S, et al. TIE2‐expressing monocytes/macrophages regulate revascularization of the ischemic limb. EMBO Mol Med. 2013;5(6):858‐869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Wolfram JA, Diaconu D, Hatala DA, et al. Keratinocyte but not endothelial cell‐specific overexpression of Tie2 leads to the development of psoriasis. Am J Pathol. 2009;174(4):1443‐1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Hillen F, Kaijzel EL, Castermans K, oude Egbrink MGA, Löwik CWGM, Griffioen AW. A transgenic Tie2‐GFP athymic mouse model; a tool for vascular biology in xenograft tumors. Biochem Biophys Res Commun. 2008;368(2):364‐367. [DOI] [PubMed] [Google Scholar]
- 70. Saeidnia S, Manayi A, Abdollahi M. From in vitro experiments to in vivo and clinical studies; pros and cons. Curr Drug Discov Technol. 2015;12(4):218‐224. [DOI] [PubMed] [Google Scholar]
- 71. Katt ME, Placone AL, Wong AD, Xu ZS, Searson PC. In vitro tumor models: advantages, disadvantages, variables, and selecting the right platform. Front Bioeng Biotechnol. 2016;4:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Holmgaard A, Askou AL, Benckendorff JNE, et al. In vivo knockout of the vegfa gene by lentiviral delivery of CRISPR/Cas9 in mouse retinal pigment epithelium cells. Mol Ther Nucleic Acids. 2017;9:89‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Crauciuc A, Tripon F, Gheorghiu A, Nemes G, Boglis A, Banescu C. Review. Development, applications, benefits, challenges and limitations of the new genome engineering technique. An update study. Acta Med Marisiensis. 2017;63(1):4‐9. [Google Scholar]
- 74. Wang LV, Gao L. Photoacoustic microscopy and computed tomography: from bench to bedside. Annu Rev Biomed Eng. 2014;16(1):155‐185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Mahmud MS, Cadotte DW, Vuong B, et al. Review of speckle and phase variance optical coherence tomography to visualize microvascular networks. J Biomed Optics. 2013;18(5):050901. [DOI] [PubMed] [Google Scholar]
- 76. Sydor AM, Czymmek KJ, Puchner EM, Mennella V. Super‐resolution microscopy: from single molecules to supramolecular assemblies. Trends Cell Biol. 2015;25(12):730‐748. [DOI] [PubMed] [Google Scholar]
- 77. Dellian M, Witwer BP, Salehi HA, Yuan F, Jain RK. Quantitation and physiological characterization of angiogenic vessels in mice: effect of basic fibroblast growth factor, vascular endothelial growth factor/vascular permeability factor, and host microenvironment. Am J Pathol. 1996;149(1):59‐71. [PMC free article] [PubMed] [Google Scholar]
- 78. Zudaire E, Gambardella L, Kurcz C, Vermeren S. A computational tool for quantitative analysis of vascular networks. PLoS ONE. 2011;6(11):e27385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Strahler AN. Quantitative analysis of watershed geomorphology. Trans Am Geophys Union. 1957;38(6):913. [Google Scholar]
- 80. Fenton BM, Zweifach BW. Microcirculatory model relating geometrical variation to changes in pressure and flow rate. Ann Biomed Eng. 1981;9(4):303‐321. [Google Scholar]
- 81. Seaman ME, Peirce SM, Kelly K. Rapid analysis of vessel elements (RAVE): a tool for studying physiologic, pathologic and tumor angiogenesis. PLoS ONE. 2011;6(6):e20807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Stamatelos SK, Kim E, Pathak AP, Popel AS. A bioimage informatics based reconstruction of breast tumor microvasculature with computational blood flow predictions. Microvasc Res. 2014;91:8‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Niemisto A, Dunmire V, Yli‐Harja O, Zhang W, Shmulevich I. Robust quantification of in vitro angiogenesis through image analysis. IEEE Trans Med Imaging. 2005;24(4):549‐553. [DOI] [PubMed] [Google Scholar]
- 84. Sové RJ, Goldman D, Fraser GM. A computational model of the effect of capillary density variability on oxygen transport, glucose uptake, and insulin sensitivity in prediabetes. Microcirculation. 2017;24(2):e12342. [DOI] [PubMed] [Google Scholar]
- 85. Nolan DJ, Ginsberg M, Israely E, et al. Molecular signatures of tissue‐specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell. 2013;26(2):204‐219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Chi J‐T, Chang HY, Haraldsen G, et al. Endothelial cell diversity revealed by global expression profiling. Proc Natl Acad Sci USA. 2003;100(19):10623‐10628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Banavar JR, Cooke TJ, Rinaldo A, Maritan A. Form, function, and evolution of living organisms. Proc Natl Acad Sci. 2014;111(9):3332‐3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Fraser GM, Goldman D, Ellis CG. Comparison of generated parallel capillary arrays to three‐dimensional reconstructed capillary networks in modeling oxygen transport in discrete microvascular volumes. Microcirculation (New York, N.Y. : 1994). 2013;20(8):748‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Fraser GM, Goldman D, Ellis CG. Microvascular flow modeling using in vivo hemodynamic measurements in reconstructed 3D capillary networks. Microcirculation (New York, N.Y. : 1994). 2012;19(6):510‐520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Davis GE, Norden PR, Bowers SLK. Molecular control of capillary morphogenesis and maturation by recognition and remodeling of the extracellular matrix: functional roles of endothelial cells and pericytes in health and disease. Connect Tissue Res. 2015;56(5):392‐402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Han H‐C. Twisted blood vessels: symptoms, etiology and biomechanical mechanisms. J Vasc Res. 2012;49(3):185‐197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Tykocki NR, Boerman EM, Jackson WF. Smooth muscle ion channels and regulation of vascular tone in resistance arteries and arterioles. Compr Physiol. 2017;7(2):485‐581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Hamilton NB, Attwell D, Hall CN. Pericyte‐mediated regulation of capillary diameter: a component of neurovascular coupling in health and disease. Front Neuroenergetics. 2010;2:1‐15. https://www.frontiersin.org/articles/10.3389/fnene.2010.00005/full [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Carroll RG. Elsevier's Integrated Physiology. Philadelphia, PA: Mosby Elsevier; 2007. [Google Scholar]
- 95. Granger DN, Senchenkova E. Inflammation and the Microcirculation. San Rafael, CA: Morgan & Claypool Life Sciences; 2010. (Integrated Systems Physiology—From Cell to Function). [PubMed] [Google Scholar]
- 96. Pfister F, Feng Y, vom Hagen F, et al. Pericyte migration: a novel mechanism of pericyte loss in experimental diabetic retinopathy. Diabetes. 2008;57(9):2495‐2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Morin KT, Carlson PD, Tranquillo RT. Automated image analysis programs for the quantification of microvascular network characteristics. Methods (San Diego, Calif.). 2015;84:76‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Ding L, Cheng R, Hu Y, et al. Peroxisome proliferator–activated receptor α protects capillary pericytes in the retina. Am J Pathol. 2014;184(10):2709‐2720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Shami A, Knutsson A, Dunér P, et al. Dystrophin deficiency reduces atherosclerotic plaque development in ApoE‐null mice. Sci Rep. 2015;5(1):13904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Seaman SA, Cao Y, Campbell CA, Peirce SM. Macrophage recruitment and polarization during collateral vessel remodeling in murine adipose tissue. Microcirculation (New York, N.Y. : 1994). 2016;23(1):75‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Corliss BA, Azimi MS, Munson JM, Peirce SM, Murfee WL. Macrophages: an inflammatory link between angiogenesis and lymphangiogenesis. Microcirculation (New York, N.Y.: 1994). 2016;23(2):95‐121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Wu J, Hadoke PW, Takov K, Korczak A, Denvir M, Smith L. Influence of androgen receptor in vascular cells on reperfusion following hindlimb ischaemia. PLoS ONE. 2016;11:e0154987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Rueden CT, Schindelin J, Hiner MC, et al. Image J2: ImageJ for the next generation of scientific image data. BMC Bioinformatics. 2017;18:529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Schindelin J, Arganda‐Carreras I, Frise E, et al. Fiji: an open‐source platform for biological‐image analysis. Nat Methods. 2012;9(7):676‐682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Manjunathan R, Ragunathan M. Chicken chorioallantoic membrane as a reliable model to evaluate osteosarcoma—an experimental approach using SaOS2 cell line. Biol Proced Online. 2015;17:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Sprindzuk M, Dmitruk A, Kovalev V, et al. Computer‐aided image processing of angiogenic histological. J Clin Med Res. 2009;1(5):249‐261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Peirce SM. Computational and mathematical modeling of angiogenesis. Microcirculation (New York, N.Y. : 1994). 2008;15(8):739‐751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Hira ZM, Gillies DF. A review of feature selection and feature extraction methods applied on microarray data. Adv Bioinformatics. 2015;2015:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Hoover AD, Kouznetsova V, Goldbaum M. Locating blood vessels in retinal images by piecewise threshold probing of a matched filter response. IEEE Trans Med Imaging. 2000;19(3):203‐210. [DOI] [PubMed] [Google Scholar]
- 110. Staal J, Abramoff MD, Niemeijer M, Viergever MA, van Ginneken B. Ridge‐based vessel segmentation in color images of the retina. IEEE Trans Med Imaging. 2004;23(4):501‐509. [DOI] [PubMed] [Google Scholar]
- 111. Goel R, Kumar V, Srivastava S, Sinha AK. A review of feature extraction techniques for image analysis. IJARCET. 2017;6(2):3. [Google Scholar]
- 112. Pries AR, Secomb TW. Making microvascular networks work: angiogenesis, remodeling, and pruning. Physiology. 2014;29(6):446‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Fry BC, Lee J, Smith NP, Secomb TW. Estimation of blood flow rates in large microvascular networks. Microcirculation. 2012;19(6):530‐538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Secomb TW. Theoretical models for regulation of blood flow. Microcirculation. 2015;15(8):765‐775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Gould IG, Linninger AA. Hematocrit distribution and tissue oxygenation in large microcirculatory networks. Microcirculation. 2015;22(1):1‐18. [DOI] [PubMed] [Google Scholar]
- 116. Kuliga KZ, McDonald EF, Gush R, Michel C, Chipperfield AJ, Clough GF. Dynamics of microvascular blood flow and oxygenation measured simultaneously in human skin. Microcirculation. 2014;21(6):562‐573. [DOI] [PubMed] [Google Scholar]
- 117. Paradowski M, Kwasnicka H, Borysewicz K. Capillary blood vessel tortuosity measurement using graph analysis In: Setchi R, Jordanov I, eds.Knowledge‐Based and Intelligent Information and Engineering Systems. Berlin, Heidelberg: Springer; 2009:135‐142. (Lecture Notes in Computer Science). [Google Scholar]
- 118. Vargas R, Waldron A, Sharma A, Flórez R, Narayan DA. A graph theoretic analysis of leverage centrality. AKCE Int J Graphs Comb. 2017;14(3):295‐306. [Google Scholar]
- 119. Di Lanzo C, Marzetti L, Zappasodi F, De Vico Fallani F, Pizzella V. Redundancy as a graph‐based index of frequency specific MEG functional connectivity. Comput Math Methods Med. 2012;2012:207305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Malliaros FD, Rossi M‐EG, Vazirgiannis M. Locating influential nodes in complex networks. Sci Rep. 2016;6:19307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Zhang W, Chien J, Yong J, Kuang R. Network‐based machine learning and graph theory algorithms for precision oncology. NPJ Precis Oncol. 2017;1(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Niepert M, Ahmed M, Kutzkov K. Learning Convolutional Neural Networks for Graphs. Proceedings of The 33rd International Conference on Machine Learning, in PMLR 48 2016:2014‐2023.
- 123. Lawrence E, Michailidis G, Nair VN, Xi B. Network tomography: a review and recent developments In: Fan J, Koul H, eds. Frontiers in Statistics. London: Imperial College Press; 2006:345‐366. [Google Scholar]
- 124. Coates A, Hero AO III, Nowak R, Yu B. Internet tomography. IEEE Signal Process Mag. 2002;19(3):47‐65. [Google Scholar]
- 125. Abramoff MD, Garvin MK, Sonka M. Retinal imaging and image analysis. IEEE Rev Biomed Eng. 2010;3:169‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Trucco E, Ruggeri A, Karnowski T, et al. Validating retinal fundus image analysis algorithms: issues and a proposal. Invest Ophthalmol Vis Sci. 2013;54(5):3546‐3559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. van der Heijden AA, Abramoff MD, Verbraak F, van Hecke MV, Liem A, Nijpels G. Validation of automated screening for referable diabetic retinopathy with the IDx‐DR device in the Hoorn Diabetes Care System. Acta Ophthalmol. 2018;96(1):63‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Batabyal T, Vaccari A, Acton ST. NeuroBFD: Size‐independent automated classification of neurons using conditional distributions of morphological features. In: 2018 IEEE 15th International Symposium on Biomedical Imaging (ISBI 2018); 2018:912‐915.
- 129. Batabyal T, Acton ST. ElasticPath2Path: Automated morphological classification of neurons by elastic path matching. arXiv:1802.06913 [eess, q‐bio]; 2018. http://arxiv.org/abs/1802.06913. Accessed August 22. 2018
- 130. Vasquez SX, Gao F, Su F, et al. Optimization of MicroCT imaging and blood vessel diameter quantitation of preclinical specimen vasculature with radiopaque polymer injection medium. PLoS ONE. 2011;6(4):e19099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Bruyninckx P, Loeckx D, Vandermeulen D, Suetens P. Segmentation of lung vessel trees by global optimization. In: Pluim JPW, Dawant BM, eds. . Proceedings of SPIE 7259, Medical ImagingLake Buena Vista, FL; 2009. [Google Scholar]
- 132. Corliss BA, Ray HC, Patrie J, et al. CIRCOAST: a statistical hypothesis test for cellular colocalization with network structures. Bioinformatics. 2018;1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Stintzing S, Ocker M, Hartner A, Amann K, Barbera L, Neureiter D. Differentiation patterning of vascular smooth muscle cells (VSMC) in atherosclerosis. Virchows Arch. 2009;455(2):171‐185. [DOI] [PubMed] [Google Scholar]
- 134. Liu L, Shi G‐P. CD31: beyond a marker for endothelial cells. Cardiovasc Res. 2012;94(1):3‐5. [DOI] [PubMed] [Google Scholar]
- 135. Yu QC, Song W, Wang D, Zeng YA. Identification of blood vascular endothelial stem cells by the expression of protein C receptor. Cell Res. 2016;26(10):1079‐1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Kumar A, D'Souza SS, Moskvin OV, et al. Specification and diversification of pericytes and smooth muscle cells from mesenchymoangioblasts. Cell Rep. 2017;19(9):1902‐1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Fang X, Djouhri L, McMullan S, et al. Intense isolectin‐B4 binding in rat dorsal root ganglion neurons distinguishes C‐fiber nociceptors with broad action potentials and high Nav1.9 expression. J Neurosci. 2006;26(27):7281‐7292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Kubo A, Katanosaka K, Mizumura K. Extracellular matrix proteoglycan plays a pivotal role in sensitization by low pH of mechanosensitive currents in nociceptive sensory neurones. J Physiol. 2012;590(13):2995‐3007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Ismail JA, Poppa V, Kemper LE, et al. Immunohistologic labeling of murine endothelium. Cardiovasc Pathol. 2003;12(2):82‐90. [DOI] [PubMed] [Google Scholar]
- 140. Zakharova IS, Zhiven’ MK, Saaya SB, et al. Endothelial and smooth muscle cells derived from human cardiac explants demonstrate angiogenic potential and suitable for design of cell‐containing vascular grafts. J Transl Med. 2017;15:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Lünemann A, Ullrich O, Diestel A, et al. Macrophage/microglia activation factor expression is restricted to lesion‐associated microglial cells after brain trauma. Glia. 2006;53(4):412‐419. [DOI] [PubMed] [Google Scholar]
- 142. Wattananit S, Tornero D, Graubardt N, et al. Monocyte‐derived macrophages contribute to spontaneous long‐term functional recovery after stroke in mice. J Neurosci. 2016;36(15):4182‐4195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Bahramsoltani M, Slosarek I, De Spiegelaere W, Plendl J. Angiogenesis and collagen type IV expression in different endothelial cell culture systems. Anat Histol Embryol. 2014;43(2):103‐115. [DOI] [PubMed] [Google Scholar]
- 144. Sava P, Cook IO, Mahal RS, Gonzalez AL. Human microvascular pericyte basement membrane remodeling regulates neutrophil recruitment. Microcirculation. 2015;22(1):54‐67. [DOI] [PubMed] [Google Scholar]
- 145. Katsuda S, Okada Y, Minamoto T, Nakanishi I. Enhanced synthesis of type IV collagen in cultured arterial smooth muscle cells associated with phenotypic modulation by dimethyl sulfoxide. Cell Biol Int Rep. 1987;11(12):861‐870. [DOI] [PubMed] [Google Scholar]
- 146. Feru J, Delobbe E, Ramont L, et al. Aging decreases collagen IV expression in vivo in the dermo‐epidermal junction and in vitro in dermal fibroblasts: possible involvement of TGF‐β1. Eur J Dermatol. 2016;26(4):350‐360. [DOI] [PubMed] [Google Scholar]
- 147. Sillat T, Saat R, Pöllänen R, Hukkanen M, Takagi M, Konttinen YT. Basement membrane collagen type IV expression by human mesenchymal stem cells during adipogenic differentiation. J Cell Mol Med. 2012;16(7):1485‐1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Kim H, Yoon CS, Kim H, Rah B. Expression of extracellular matrix components fibronectin and laminin in the human fetal heart. Cell Struct Funct. 1999;24(1):19‐26. [DOI] [PubMed] [Google Scholar]
- 149. Yousif LF, Di Russo J, Sorokin L. Laminin isoforms in endothelial and perivascular basement membranes. Cell Adh Migr. 2013;7(1):101‐110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Godin LM, Sandri BJ, Wagner DE, et al. Decreased laminin expression by human lung epithelial cells and fibroblasts cultured in acellular lung scaffolds from aged mice. PLoS ONE. 2016;11(3):e0150966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Ekblom M, Falk M, Salmivirta K, Durbeej M, Ekblom P. Laminin isoforms and epithelial development. Ann N Y Acad Sci. 1998;857:194‐211. [DOI] [PubMed] [Google Scholar]
- 152. Fina L, Molgaard HV, Robertson D, et al. Expression of the CD34 gene in vascular endothelial cells. Blood. 1990;75(12):2417‐2426. [PubMed] [Google Scholar]
- 153. Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193‐215. [DOI] [PubMed] [Google Scholar]
- 154. Hindle P, Khan N, Biant L, Péault B. The infrapatellar fat pad as a source of perivascular stem cells with increased chondrogenic potential for regenerative medicine. Stem Cells Transl Med. 2017;6(1):77‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Sidney LE, Branch MJ, Dunphy SE, Dua HS, Hopkinson A. Concise review: evidence for CD34 as a common marker for diverse progenitors. Stem Cells (Dayton, Ohio). 2014;32(6):1380‐1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Giampietro C, Taddei A, Corada M, et al. Overlapping and divergent signaling pathways of N‐cadherin and VE‐cadherin in endothelial cells. Blood. 2012;119(9):2159‐2170. [DOI] [PubMed] [Google Scholar]
- 157. Chang L, Noseda M, Higginson M, et al. Differentiation of vascular smooth muscle cells from local precursors during embryonic and adult arteriogenesis requires Notch signaling. Proc Natl Acad Sci. 2012;109(18):6993‐6998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Gavard J. Endothelial permeability and VE‐cadherin. Cell Adh Migr. 2014;8(2):158‐164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Zahr A, Alcaide P, Yang J, et al. Endomucin prevents leukocyte–endothelial cell adhesion and has a critical role under resting and inflammatory conditions. Nat Commun. 2016;7:10363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Liu C, Shao ZM, Zhang L, et al. Human endomucin is an endothelial marker. Biochem Biophys Res Commun. 2001;288(1):129‐136. [DOI] [PubMed] [Google Scholar]
- 161. Samulowitz U, Kuhn A, Brachtendorf G, et al. Human endomucin. Am J Pathol. 2002;160(5):1669‐1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Brachtendorf G, Kuhn A, Samulowitz U, et al. Early expression of endomucin on endothelium of the mouse embryo and on putative hematopoietic clusters in the dorsal aorta. Dev Dyn. 2001;222(3):410‐419. [DOI] [PubMed] [Google Scholar]
- 163. Zanetta L, Marcus SG, Vasile J, et al. Expression of Von Willebrand factor, an endothelial cell marker, is up‐regulated by angiogenesis factors: a potential method for objective assessment of tumor angiogenesis. Int J Cancer. 2000;85(2):281‐288. [DOI] [PubMed] [Google Scholar]
- 164. Trost A, Lange S, Schroedl F, et al. Brain and retinal pericytes: origin, function and role. Front Cell Neurosci. 2016;10:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Balabanov R, Dore‐Duffy P. Role of the CNS microvascular pericyte in the blood‐brain barrier. J Neurosci Res. 1998;53(6):637‐644. [DOI] [PubMed] [Google Scholar]
- 166. Strauss O, Phillips A, Ruggiero K, Bartlett A, Dunbar PR. Immunofluorescence identifies distinct subsets of endothelial cells in the human liver. Sci Rep. 2017;7:44356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Bergers G, Song S. The role of pericytes in blood‐vessel formation and maintenance. Neuro Oncol. 2005;7(4):452‐464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Merkulova‐Rainon T, Broqueres‐You D, Kubis N, Silvestre J‐S, Levy BI. Towards the therapeutic use of vascular smooth muscle progenitor cells. Cardiovasc Res. 2012;95(2):205‐214. [DOI] [PubMed] [Google Scholar]
- 169. Hinz B, Celetta G, Tomasek JJ, Gabbiani G, Chaponnier C. Alpha‐smooth muscle actin expression upregulates fibroblast contractile activity. Mol Biol Cell. 2001;12(9):2730‐2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Lindskog H, Athley E, Larsson E, Lundin S, Hellström M, Lindahl P. New insights to vascular smooth muscle cell and pericyte differentiation of mouse embryonic stem cells in vitro. Arterioscler Thromb Vasc Biol. 2006;26(7):1457‐1464. [DOI] [PubMed] [Google Scholar]
- 171. Haskins RM, Nguyen AT, Alencar GF, et al. Klf4 has an unexpected protective role in perivascular cells within the microvasculature. Am J Physiol Heart Circ Physiol. 2018;315:H402‐H414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Shankman LS, Gomez D, Cherepanova OA, et al. KLF4 dependent phenotypic modulation of SMCs plays a key role in atherosclerotic plaque pathogenesis. Nat Med. 2015;21(6):628‐637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Fukushi J, Makagiansar IT, Stallcup WB. NG2 proteoglycan promotes endothelial cell motility and angiogenesis via engagement of galectin‐3 and α3β1 integrin. Mol Biol Cell. 2004;15(8):3580‐3590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Zhu L, Xiang P, Guo K, et al. Microglia/monocytes with NG2 expression have no phagocytic function in the cortex after LPS focal injection into the rat brain. Glia. 2012;60(9):1417‐1426. [DOI] [PubMed] [Google Scholar]
- 175. Yan X, Yan L, Liu S, Shan Z, Tian Y, Jin Z. N‐cadherin, a novel prognostic biomarker, drives malignant progression of colorectal cancer. Mol Med Rep. 2015;12(2):2999‐3006. [DOI] [PubMed] [Google Scholar]
- 176. Tsuchiya B, Sato Y, Kameya T, Okayasu I, Mukai K. Differential expression of N‐cadherin and E‐cadherin in normal human tissues. Arch Histol Cytol. 2006;69(2):135‐145. [DOI] [PubMed] [Google Scholar]
- 177. Sun Z, Parrish AR, Hill MA, Meininger GA. N‐cadherin, a vascular smooth muscle cell‐cell adhesion molecule: function and signaling for vasomotor control. Microcirculation. 2014;21(3):208‐218. [DOI] [PubMed] [Google Scholar]
- 178. Ishimine H, Yamakawa N, Sasao M, et al. N‐Cadherin is a prospective cell surface marker of human mesenchymal stem cells that have high ability for cardiomyocyte differentiation. Biochem Biophys Res Commun. 2013;438(4):753‐759. [DOI] [PubMed] [Google Scholar]
- 179. Council L, Hameed O. Differential expression of immunohistochemical markers in bladder smooth muscle and myofibroblasts, and the potential utility of desmin, smoothelin, and vimentin in staging of bladder carcinoma. Mod Pathol. 2009;22(5):639‐650. [DOI] [PubMed] [Google Scholar]
- 180. Gerhardt H, Betsholtz C. Endothelial‐pericyte interactions in angiogenesis. Cell Tissue Res. 2003;314(1):15‐23. [DOI] [PubMed] [Google Scholar]
- 181. Hewitt KJ, Shamis Y, Knight E, et al. PDGFRβ expression and function in fibroblasts derived from pluripotent cells is linked to DNA demethylation. J Cell Sci. 2012;125(9):2276‐2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Lindblom P, Gerhardt H, Liebner S, et al. Endothelial PDGF‐B retention is required for proper investment of pericytes in the microvessel wall. Genes Dev. 2003;17(15):1835‐1840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Damisah EC, Hill RA, Tong L, Murray KN, Grutzendler J. A fluoro‐Nissl dye identifies pericytes as distinct vascular mural cells during in vivo brain imaging. Nat Neurosci. 2017;20(7):1023‐1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Hill M, Davis M. Local Control of Microvascular Perfusion. San Rafael, CA: Morgan & Claypool Publishers; 2012. [Google Scholar]
- 185. Clemetson KJ, Clemetson JM. Platelet receptors A2 In: Michelson AD, ed. Platelets, 3rd edn San Diego, CA: Academic Press; 2013:169‐194. [Google Scholar]
- 186. Fukuhara S, Sako K, Noda K, Nagao K, Miura K, Mochizuki N. Tie2 is tied at the cell‐cell contacts and to extracellular matrix by Angiopoietin‐1. Exp Mol Med. 2009;41(3):133‐139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Ito K, Turcotte R, Cui J, et al. Self‐renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science (New York, N.Y.). 2016;354(6316):1156‐1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Li S, Li T, Luo Y, et al. Retro‐orbital injection of FITC‐dextran is an effective and economical method for observing mouse retinal vessels. Mol Vis. 2011;17:3566‐3573. [PMC free article] [PubMed] [Google Scholar]
- 189. Roberts JJ, Martens PJ. Engineering biosynthetic cell encapsulation systems In: Poole‐Warren L, Martens P, Green R, eds. Biosynthetic Polymers for Medical Applications. London: Woodhead Publishing; 2016:205‐239. (Woodhead Publishing Series in Biomaterials). [Google Scholar]
- 190. Kappel A, Rönicke V, Damert A, Flamme I, Risau W, Breier G. Identification of vascular endothelial growth factor (VEGF) receptor‐2 (Flk‐1) promoter/enhancer sequences sufficient for angioblast and endothelial cell‐specific transcription in transgenic mice. Blood. 1999;93(12):4284‐4292. [PubMed] [Google Scholar]
- 191. Haruta H, Nagata Y, Todokoro K. Role of Flk‐1 in mouse hematopoietic stem cells. FEBS Lett. 2001;507(1):45‐48. [DOI] [PubMed] [Google Scholar]
- 192. Choi J‐S, Kim H‐Y, Cha J‐H, et al. Upregulation of vascular endothelial growth factor receptors Flt‐1 and Flk‐1 following acute spinal cord contusion in rats. J Histochem Cytochem. 2007;55(8):821‐830. [DOI] [PubMed] [Google Scholar]
- 193. Yang S‐Z, Zhang L‐M, Huang Y‐L, Sun F‐Y. Distribution of Flk‐1 and Flt‐1 receptors in neonatal and adult rat brains. Anat Rec A Discov Mol Cell Evol Biol. 2003;274(1):851‐856. [DOI] [PubMed] [Google Scholar]
- 194. Miettinen M, Rikala M‐S, Rysz J, Lasota J, Wang Z‐F. Vascular endothelial growth factor receptor 2 (VEGFR2) as a marker for malignant vascular tumors and mesothelioma – immunohistochemical study of 262 vascular endothelial and 1640 nonvascular tumors. Am J Surg Pathol. 2012;36(4):629‐639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Eilken HM, Diéguez‐Hurtado R, Schmidt I, et al. Pericytes regulate VEGF‐induced endothelial sprouting through VEGFR1. Nat Commun. 2017;8(1):1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Motoike T, Loughna S, Perens E, et al. Universal GFP reporter for the study of vascular development. Genesis (New York, N.Y.: 2000). 2000;28(2):75‐81. [DOI] [PubMed] [Google Scholar]
- 197. Cha YR, Weinstein BM. Visualization and experimental analysis of blood vessel formation using transgenic zebrafish. Birth Defects Res C Embryo Today. 2007;81(4):286‐296. [DOI] [PubMed] [Google Scholar]
- 198. Kisanuki YY, Hammer RE, Miyazaki J, Williams SC, Richardson JA, Yanagisawa M. Tie2‐Cre transgenic mice: a new model for endothelial cell‐lineage analysis in vivo. Dev Biol. 2001;230(2):230‐242. [DOI] [PubMed] [Google Scholar]
- 199. Constien R, Forde A, Liliensiek B, et al. Characterization of a novel EGFP reporter mouse to monitor Cre recombination as demonstrated by a Tie2 Cre mouse line. Genesis (New York, N.Y.: 2000). 2001;30(1):36‐44. [DOI] [PubMed] [Google Scholar]
- 200. Forde A, Constien R, Gröne H‐J, Hämmerling G, Arnold B. Temporal Cre‐mediated recombination exclusively in endothelial cells using Tie2 regulatory elements. Genesis. 2002;33(4):191‐197. [DOI] [PubMed] [Google Scholar]
- 201. Hartmann DA, Underly RG, Watson AN, Shih AY. A murine toolbox for imaging the neurovascular unit. Microcirculation (New York, N.Y. : 1994). 2015;22(3):168‐182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Alva JA, Zovein AC, Monvoisin A, et al. VE‐Cadherin‐Cre‐recombinase transgenic mouse: a tool for lineage analysis and gene deletion in endothelial cells. Dev Dyn. 2006;235(3):759‐767. [DOI] [PubMed] [Google Scholar]
- 203. Chen Q, Zhang H, Liu Y, et al. Endothelial cells are progenitors of cardiac pericytes and vascular smooth muscle cells. Nat Commun. 2016;7:12422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Zhong W, Gao X, Wang S, et al. Prox1‐GFP/Flt1‐DsRed transgenic mice: an animal model for simultaneous live imaging of angiogenesis and lymphangiogenesis. Angiogenesis. 2017;20(4):581‐598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Lawson ND, Weinstein BM. In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol. 2002;248(2):307‐318. [DOI] [PubMed] [Google Scholar]
- 206. Poché RA, Larina IV, Scott ML, Saik JE, West JL, Dickinson ME. The Flk1‐myr:mCherry mouse as a useful reporter to characterize multiple aspects of ocular blood vessel development and disease. Dev Dyn. 2009;238(9):2318‐2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Jin S‐W, Herzog W, Santoro MM, et al. A transgene‐assisted genetic screen identifies essential regulators of vascular development in vertebrate embryos. Dev Biol. 2007;307(1):29‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Larina IV, Shen W, Kelly OG, Hadjantonakis A‐K, Baron MH, Dickinson ME. A membrane associated mCherry fluorescent reporter line for studying vascular remodeling and cardiac function during murine embryonic development. Anat Rec (Hoboken, N.J.: 2007). 2009;292(3):333‐341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Cuervo H, Pereira B, Nadeem T, et al. PDGFRβ‐P2A‐CreERT2 mice: a genetic tool to target pericytes in angiogenesis. Angiogenesis. 2017;20(4):655‐662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Zhu X, Bergles DE, Nishiyama A. NG2 cells generate both oligodendrocytes and gray matter astrocytes. Development. 2008;135(1):145‐157. [DOI] [PubMed] [Google Scholar]
- 211. Volz KS, Jacobs AH, Chen HI, et al. Pericytes are progenitors for coronary artery smooth muscle. eLife. 2015;4:1‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Iljin K, Petrova TV, Veikkola T, Kumar V, Poutanen M, Alitalo K. A fluorescent Tie1 reporter allows monitoring of vascular development and endothelial cell isolation from transgenic mouse embryos. FASEB J. 2002;16(13):1764‐1774. [DOI] [PubMed] [Google Scholar]
- 213. Claxton S, Kostourou V, Jadeja S, Chambon P, Hodivala‐Dilke K, Fruttiger M. Efficient, inducible Cre‐recombinase activation in vascular endothelium. Genesis (New York, N.Y.: 2000). 2008;46(2):74‐80. [DOI] [PubMed] [Google Scholar]
- 214. Fernández‐Klett F, Offenhauser N, Dirnagl U, Priller J, Lindauer U. Pericytes in capillaries are contractile in vivo, but arterioles mediate functional hyperemia in the mouse brain. Proc Natl Acad Sci USA. 2010;107(51):22290‐22295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Proebstl D, Voisin M‐B, Woodfin A, et al. Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J Exp Med. 2012;209(6):1219‐1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Yokota T, Kawakami Y, Nagai Y, et al. Bone marrow lacks a transplantable progenitor for smooth muscle type alpha‐actin‐expressing cells. Stem Cells (Dayton, Ohio). 2006;24(1):13‐22. [DOI] [PubMed] [Google Scholar]
- 217. Wendling O, Bornert J‐M, Chambon P, Metzger D. Efficient temporally‐controlled targeted mutagenesis in smooth muscle cells of the adult mouse. Genesis (New York, N.Y.: 2000). 2009;47(1):14‐18. [DOI] [PubMed] [Google Scholar]
- 218. Nguyen M, Shing Y, Folkman J. Quantitation of angiogenesis and antiangiogenesis in the chick embryo chorioallantoic membrane. Microvasc Res. 1994;47(1):31‐40. [DOI] [PubMed] [Google Scholar]
- 219. Gavins FNE, Chatterjee BE. Intravital microscopy for the study of mouse microcirculation in anti‐inflammatory drug research: focus on the mesentery and cremaster preparations. J Pharmacol Toxicol Methods. 2004;49(1):1‐14. [DOI] [PubMed] [Google Scholar]
- 220. Boerman EM, Segal SS. Depressed perivascular sensory innervation of mouse mesenteric arteries with advanced age. J Physiol. 2016;594(8):2323‐2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Lipowsky HH, Zweifach BW. Network analysis of microcirculation of cat mesentery. Microvasc Res. 1974;7(1):73‐83. [DOI] [PubMed] [Google Scholar]
- 222. Sinkler SY, Segal SS. Aging alters reactivity of microvascular resistance networks in mouse gluteus maximus muscle. Am J Physiol Heart Circ Physiol. 2014;307(6):H830‐H839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Socha MJ, Segal SS. Isolation of microvascular endothelial tubes from mouse resistance arteries. J Vis Exp. 2013;81:e50759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Jackson WF, Huebner JM, Rusch NJ. Enzymatic isolation and characterization of single vascular smooth muscle cells from cremasteric arterioles. Microcirculation (New York, N.Y.: 1994). 1997;4(1):35‐50. [DOI] [PubMed] [Google Scholar]
- 225. Asai A, Sahani N, Ouchi Y, Martyn J, Yasuhara S. In vivo micro‐circulation measurement in skeletal muscle by intra‐vital microscopy. J Vis Exp. 2007;4:210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Menth‐Chiari WA, Curl WW, Rosencrance E, Smith TL. Contusion of skeletal muscle increases leukocyte‐endothelial cell interactions: an intravital‐microscopy study in rats. J Trauma. 1998;45(4):709‐714. [DOI] [PubMed] [Google Scholar]
- 227. Bagher P, Segal SS. The mouse cremaster muscle preparation for intravital imaging of the microcirculation. J Vis Exp. 2011;(52): 10.3791/2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Gaber MW, Yuan H, Killmar JT, Naimark MD, Kiani MF, Merchant TE. An intravital microscopy study of radiation‐induced changes in permeability and leukocyte–endothelial cell interactions in the microvessels of the rat pia mater and cremaster muscle. Brain Res Protoc. 2004;13(1):1‐10. [DOI] [PubMed] [Google Scholar]
- 229. Eisen JS, Smith JC. Controlling morpholino experiments: don't stop making antisense. Development. 2008;135(10):1735‐1743. [DOI] [PubMed] [Google Scholar]
- 230. Coonrod SA, Bolling LC, Wright PW, Visconti PE, Herr JC. A morpholino phenocopy of the mouse MOS mutation. Genesis. 2001;30(3):198‐200. [DOI] [PubMed] [Google Scholar]
- 231. Clark ER. Studies on the growth of blood‐vessels in the tail of the frog larva?by observation and experiment on the living animal. Am J Anat. 1918;23(1):37‐88. [Google Scholar]
- 232. Kelly‐Goss MR, Ning B, Bruce AC, et al. Dynamic, heterogeneous endothelial Tie2 expression and capillary blood flow during microvascular remodeling. Sci Rep. 2017;7(1):9049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. van Duinen V, Trietsch SJ, Joore J, Vulto P, Hankemeier T. Microfluidic 3D cell culture: from tools to tissue models. Curr Opin Biotechnol. 2015;35:118‐126. [DOI] [PubMed] [Google Scholar]
- 234. Sánchez‐Palencia DM, Bigger‐Allen A, Saint‐Geniez M, Arboleda‐Velásquez JF, D'Amore PA. Coculture assays for endothelial cells‐mural cells interactions. Methods Mol Biol (Clifton, N.J.). 2016;1464:35‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Waters J, Kluger M, Graham M, Chang W, Bradley J, Pober J. In vitro self‐assembly of human pericyte‐supported endothelial microvessels in three‐dimensional co‐culture: a simple model for interrogating endothelial:pericyte interactions. J Vascular Res. 2013;50(4):324‐331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Stahl A, Connor KM, Sapieha P, et al. The mouse retina as an angiogenesis model. Invest Ophthalmol Vis Sci. 2010;51(6):2813‐2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Jackson M, Taylor AH, Jones EA, Forrester LM. The culture of mouse embryonic stem cells and formation of embryoid bodies. Methods Mol Biol (Clifton, N.J.). 2010;633:1‐18. [DOI] [PubMed] [Google Scholar]
- 238. Humpel C. Organotypic brain slice cultures: a review. Neuroscience. 2015;305:86‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Johnson TV, Martin KR. Development and characterization of an adult retinal explant organotypic tissue culture system as an in vitro intraocular stem cell transplantation model. Invest Ophthalmol Vis Sci. 2008;49(8):3503‐3512. [DOI] [PubMed] [Google Scholar]
- 240. Sawamiphak S, Ritter M, Acker‐Palmer A. Preparation of retinal explant cultures to study ex vivo tip endothelial cell responses. Nat Protoc. 2010;5(10):1659‐1665. [DOI] [PubMed] [Google Scholar]
- 241. Bull N, Johnson T, Martin K. Organotypic explant culture of adult rat retina for in vitro investigations of neurodegeneration, neuroprotection and cell transplantation; 2011. https://www.nature.com/protocolexchange/protocols/2042. Accessed July 20, 2018.
- 242. Downs KM, Temkin R, Gifford S, McHugh J. Study of the murine allantois by allantoic explants. Dev Biol. 2001;233(2):347‐364. [DOI] [PubMed] [Google Scholar]
- 243. Nakatsu MN, Hughes CCW. An optimized three‐dimensional in vitro model for the analysis of angiogenesis. Methods Enzymol. 2008;443:65‐82. [DOI] [PubMed] [Google Scholar]
- 244. Huang B, Babcock H, Zhuang X. Breaking the diffraction barrier: super‐resolution imaging of cells. Cell. 2010;143(7):1047‐1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Thorn K. A quick guide to light microscopy in cell biology. Mol Biol Cell. 2016;27(2):219‐222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Heintzmann R, Ficz G. Breaking the resolution limit in light microscopy. Brief Funct Genomics. 2006;5(4):289‐301. [DOI] [PubMed] [Google Scholar]
- 247. Fernández‐Suárez M, Ting AY. Fluorescent probes for super‐resolution imaging in living cells. Nat Rev Mol Cell Biol. 2008;9(12):929‐943. [DOI] [PubMed] [Google Scholar]
- 248. Graf BW, Boppart SA. Imaging and analysis of three‐dimensional cell culture models. Methods Mol Biol (Clifton, N.J.). 2010;591:211‐227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Jonkman J, Brown CM. Any way you slice it—a comparison of confocal microscopy techniques. J Biomol Tech. 2015;26(2):54‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Carter M, Shieh J. Microscopy In: Carter M, Shieh J, eds. Guide to Research Techniques in Neuroscience, 2nd edn San Diego, CA: Academic Press; 2015:117‐144. [Google Scholar]
- 251. Helmchen F, Denk W. Deep tissue two‐photon microscopy. Nat Methods. 2005;2(12):932‐940. [DOI] [PubMed] [Google Scholar]
- 252. Patterson GH, Piston DW. Photobleaching in two‐photon excitation microscopy. Biophys J . 2000;78(4):2159‐2162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Xia J, Yao J, Wang LV. Photoacoustic tomography: principles and advances. Electromagn Waves (Cambridge, Mass.). 2014;147:1‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Wang L, Maslov K, Xing W, Garcia‐Uribe A, Wang LV. Video‐rate functional photoacoustic microscopy at depths. J Biomed Optics. 2012;17(10):106007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Xu M, Wang LV. Photoacoustic imaging in biomedicine. Rev Sci Instrum. 2006;77(4):041101. [Google Scholar]
- 256. Rajan V, Varghese B, van Leeuwen TG, Steenbergen W. Review of methodological developments in laser Doppler flowmetry. Lasers Med Sci. 2009;24(2):269‐283. [DOI] [PubMed] [Google Scholar]
- 257. Imaging Won R. Mapping blood flow. Nat Photonics. 2011;5(7):393. [Google Scholar]
- 258. Senarathna J, Rege A, Li N, Thakor NV. Laser speckle contrast imaging: theory, instrumentation and applications. IEEE Rev Biomed Eng. 2013;6:99‐110. [DOI] [PubMed] [Google Scholar]
- 259. Zeller‐Plumhoff B, Roose T, Clough GF, Schneider P. Image‐based modelling of skeletal muscle oxygenation. J R Soc Interface. 2017;14(127):20160992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Gnyawali SC, Blum K, Pal D, et al. Retooling laser speckle contrast analysis algorithm to enhance non‐invasive high resolution laser speckle functional imaging of cutaneous microcirculation. Sci Rep. 2017;7:41048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Campbell KR, Chaudhary R, Handel JM, Patankar MS, Campagnola PJ. Polarization‐resolved second harmonic generation imaging of human ovarian cancer. J Biomed Optics. 2018;23(6):066501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Campagnola P. Second harmonic generation imaging microscopy: applications to diseases diagnostics. Anal Chem. 2011;83(9):3224‐3231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Cox G, Sheppard CJR. Practical limits of resolution in confocal and non‐linear microscopy. Microsc Res Tech. 2004;63(1):18‐22. [DOI] [PubMed] [Google Scholar]
- 264. Vuillemin N, Mahou P, Débarre D, et al. Efficient second‐harmonic imaging of collagen in histological slides using Bessel beam excitation. Sci Rep. 2016;6:29863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Bueno JM, Ávila FJ, Artal P. Comparison of second harmonic microscopy images of collagen‐based ocular tissues with 800 and 1045 nm. Biomed Opt Express. 2017;8(11):5065‐5074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Fujimoto JG. Optical coherence tomography for ultrahigh resolution in vivo imaging. Nat Biotechnol. 2003;21(11):1361‐1367. [DOI] [PubMed] [Google Scholar]
- 267. Mazlin V, Xiao P, Dalimier E, et al. In vivo imaging through the entire thickness of human cornea by full‐field optical coherence tomography. In: Ophthalmic Technologies XXVIII. Vol. 10474. International Society for Optics and Photonics; 2018:104740S https://www.spiedigitallibrary.org/conference-proceedings-of-spie/10474/104740S/In-vivo-imaging-through-the-entire-thickness-of-human-cornea/10.1117/12.2288947.short. Accessed July 30, 2018. [Google Scholar]
- 268. de Amorim Garcia Filho CA, Yehoshua Z, Gregori G, Puliafito CA, Rosenfeld PJ. Optical coherence tomography In: Ryan SJ, Sadda SR, Hinton DR, Schachat AP, Sadda SR, Wilkinson CP, Wiedemann P, Schachat AP, eds. Retina, 5th edn London: W.B. Saunders; 2013:82‐110. [Google Scholar]
- 269. Tan ACS, Tan GS, Denniston AK, et al. An overview of the clinical applications of optical coherence tomography angiography. Eye. 2018;32(2):262‐286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Watanabe S, Punge A, Hollopeter G, et al. Protein localization in electron micrographs using fluorescence nanoscopy. Nat Methods. 2011;8(1):80‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Wu R, Zhan Q, Liu H, Wen X, Wang B, He S. Optical depletion mechanism of upconverting luminescence and its potential for multi‐photon STED‐like microscopy. Opt Express. 2015;23(25):32401. [DOI] [PubMed] [Google Scholar]
- 272. Ramachandra R, de Jonge N. Optimized deconvolution for maximum axial resolution in three‐dimensional aberration‐corrected scanning transmission electron microscopy. Microsc Microanal. 2012;18(1):218‐228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Trépout S, Bastin P, Marco S. Preparation and observation of thick biological samples by scanning transmission electron tomography. J Vis Exp. 2017;121:e55215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Kanemaru T, Hirata K, Takasu S, et al. A fluorescence scanning electron microscope. Ultramicroscopy. 2009;109(4):344‐349. [DOI] [PubMed] [Google Scholar]
- 275. Reynaud EG, Kržič U, Greger K, Stelzer EHK. Light sheet‐based fluorescence microscopy: more dimensions, more photons, and less photodamage. HFSP J. 2008;2(5):266‐275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Colombelli J, Lorenzo C. Light sheet fluorescence microscopy applications for multicellular systems In: Cornea A, Conn PM, eds. Fluorescence Microscopy. Boston, MA: Academic Press; 2014:109‐120. [Google Scholar]
- 277. Doukas CN, Maglogiannis I, Chatziioannou AA. Computer‐supported angiogenesis quantification using image analysis and statistical averaging. IEEE Trans Inf Technol Biomed. 2008;12(5):650‐657. [DOI] [PubMed] [Google Scholar]
- 278. Vickerman MB, Keith PA, McKay TL, et al. VESGEN 2D: automated, user‐interactive software for quantification and mapping of angiogenic and lymphangiogenic trees and networks. Anat Rec. 2009;292(3):320‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Palachanis D, Szabó A, Merks RMH. Particle‐based simulation of ellipse‐shaped particle aggregation as a model for vascular network formation. Comput Part Mech. 2015;2(4):371‐379. [Google Scholar]
- 280. Walpole J, Mac Gabhann F, Peirce SM, Chappell JC. Agent‐based computational model of retinal angiogenesis simulates microvascular network morphology as a function of pericyte coverage. Microcirculation (New York, N.Y.: 1994). 2017;24(8); 10.1111/micc.12393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Shirinifard A, Gens JS, Zaitlen BL, Popławski NJ, Swat M, Glazier JA. 3D multi‐cell simulation of tumor growth and angiogenesis. PLoS ONE. 2009;4(10); e7190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Macklin P, McDougall S, Anderson ARA, Chaplain MAJ, Cristini V, Lowengrub J. Multiscale modelling and nonlinear simulation of vascular tumour growth. J Math Biol. 2009;58(4–5):765‐798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Kleinstreuer N, Dix D, Rountree M, et al. A computational model predicting disruption of blood vessel development. PLoS Comput Biol. 2013;9(4):e1002996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Norton K‐A, Popel AS. Effects of endothelial cell proliferation and migration rates in a computational model of sprouting angiogenesis. Sci Rep. 2016;6:36992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Moreira‐Soares M, Coimbra R, Rebelo L, Carvalho J, D M Travasso R. Angiogenic factors produced by hypoxic cells are a leading driver of anastomoses in sprouting angiogenesis–a computational study. Sci Rep. 2018;8:8726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Travasso RDM, Corvera Poiré E, Castro M, Rodrguez‐Manzaneque JC, Hernández‐Machado A. Tumor angiogenesis and vascular patterning: a mathematical model. PLoS ONE. 2011;6(5):e19989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Vilanova G, Colominas I, Gomez H. Coupling of discrete random walks and continuous modeling for three‐dimensional tumor‐induced angiogenesis. Comput Mech. 2014;53(3):449‐464. [Google Scholar]
- 288. Vilanova G, Colominas I, Gomez H. A mathematical model of tumour angiogenesis: growth, regression and regrowth. J R Soc Interface. 2017;14(126):20160918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Guerra MMDSQ, Travasso RDM. Novel approach to vascular network modeling in 3D. In: 2012 IEEE 2nd Portuguese Meeting in Bioengineering (ENBENG); 2012:1‐6.
- 290. Secomb TW, Alberding JP, Hsu R, Dewhirst MW, Pries AR. Angiogenesis: an adaptive dynamic biological patterning problem. PLoS Comput Biol. 2013;9(3):e1002983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Santagiuliana R, Ferrari M, Schrefler BA. Simulation of angiogenesis in a multiphase tumor growth model. Comput Methods Appl Mech Eng. 2016;304:197‐216. [Google Scholar]
- 292. Soltani M, Chen P. Numerical modeling of interstitial fluid flow coupled with blood flow through a remodeled solid tumor microvascular network. PLoS ONE. 2013;8(6):e67025. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials