Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jun 11.
Published in final edited form as: Circ Res. 2021 Jun 10;128(12):1927–1943. doi: 10.1161/CIRCRESAHA.121.318265

Bench-to-Bedside in Vascular Medicine: Optimizing the translational pipeline for patients with peripheral artery disease

Tom Alsaigh 1, Belinda A Di Bartolo 2, Jocelyne Mulangala 3, Gemma A Figtree 2, Nicholas J Leeper 1
PMCID: PMC8208504  NIHMSID: NIHMS1701247  PMID: 34110900

Abstract

Peripheral arterial disease (PAD) is a growing worldwide problem with a wide spectrum of clinical severity and is projected to consume more than $21 billion per year in the United States alone1. While vascular researchers have brought several therapies to the clinic in recent years2, 3, few of these approaches have leveraged advances in high-throughput discovery screens, novel translational models, or innovative trial designs. In the following review, we discuss recent advances in unbiased genomics and broader “omics” technology platforms, along with preclinical vascular models designed to enhance our understanding of disease pathobiology and prioritize targets for additional investigation. Further, we summarize novel approaches to clinical studies in subjects with claudication and ischemic ulceration, with an emphasis on streamlining and accelerating bench-to-bedside translation. By providing a framework designed to enhance each aspect of future clinical development programs, we hope to enrich the pipeline of therapies that may prevent loss of life and limb for those suffering from PAD.

Keywords: vascular biology, peripheral arterial disease, translational, atherosclerosis, genomics, angiogenesis, Peripheral Vascular Disease, Translational Studies, Vascular Disease

Introduction:

Over 200 million people worldwide have peripheral arterial disease (PAD), commonly defined as atherosclerosis involving arteries outside of the coronary circulation4. The high incidence of lower extremity PAD places an enormous burden on healthcare systems worldwide, necessitating novel strategies to curb disease progression5. Affected patients suffer from significant morbidities, including diminished blood supply to the limbs leading to claudication, critical limb ischemia, ulcer development, gangrene, and ultimately limb loss. While PAD treatments aimed at preventing progression of atherosclerosis through targeting dyslipidemia6, hypertension7 and dysregulated coagulation8 have saved lives and reduced systemic complications, identification of novel targets against disease sequelae are lacking. In addition, there likely remains a substantial group of patients whose susceptibility to atherosclerosis in their peripheral arteries is disproportionate to their risk factor burden in other vascular beds, and those who continue to rapidly progress despite best evidence-based management. To address these issues, it is of the utmost importance to develop therapies of direct relevance to patients suffering from PAD, including those with variable responses to therapy or clinical trajectories.

Here we explore the impact of strategic research collaboration across the whole translational pipeline to bring new medical solutions for patients with PAD. Building large cohorts of biobanked tissue and blood specimens will provide enhanced opportunity for discovering new mechanisms of disease susceptibility9, augmented by high throughput “omics” platforms, and massive advances in bioinformatics10. Ensuring that discoveries made through such approaches inform in vitro and in vivo models will be key to translating knowledge of disease-promoting pathways into new therapies for patients suffering from vascular disease. We begin by discussing advances and discoveries from such unbiased approaches, and opportunities for the future in regard to target identification and prioritization.

Discovery: Target Identification and Prioritization through Genomics

Unraveling the genetic architecture of vascular disease is fundamental in the effort to innovate and realize novel targets for therapeutic intervention, as well as markers of risk. Translational genomics is perhaps best exemplified by the discovery of PCSK9 as a molecular target to lower LDL cholesterol1113, paving the way to explore alternatives to statin therapy for atherosclerosis, a continued top cause of morbidity and mortality worldwide despite vast efforts to curb its deleterious effects14, 15. As was the case with PCSK9, uncovering novel therapeutic targets depends upon utilization of next-generation genomics approaches to disentangle disease complexity and home in on key drivers of disease progression. Here we explore some of the genomics approaches used to study vascular disease in an effort to guide the translational process from target identification to clinical trials.

Genome wide association studies:

Deposition of genetic information into large biobanks and databases has enabled genome wide association studies (GWAS) that have richly enhanced our understanding of inherited variants which contribute to complex disease phenotypes. Notably, GWAS have highlighted biological processes in the vessel wall and enabled identification of 9p21 as an impactful genetic locus in both coronary artery disease16, 17 and PAD18. Importantly, this locus contains the cyclin-dependent kinase inhibitor CDKN2B which was functionally shown to be involved in PAD pathogenesis after Cdkn2b−/− mice developed advanced hindlimb ischemia after femoral artery ligation19. The Million Veteran Program (MVP)20, a database established in 2011 of genomic data from the Veterans Affairs Healthcare System, is one such archive used to explore PAD susceptibility loci21. The MVP cohort includes a multiethnic population that is particularly susceptible to PAD and its comorbid sequelae due to the higher prevalence of smoking22, diabetes and hypertension23, making this an ideal group from which to identify genetic factors that increase PAD risk. Indeed, Klarin, et al utilized the MVP dataset and identified 19 total PAD loci that exceeded genome-wide significance21. Importantly, their analysis highlighted 11 genetic loci common to coronary, cerebral and peripheral vascular disease (LDLR, LPA, LPL, SORT1 – lipids; PTPN11 – hypertension; TCF7L2 – diabetes), as well as four genetic signals specific to PAD, including Factor V Leiden variant, F5p.R506Q (thrombosis) and CHRNA3 (smoking), suggesting that smoking and thrombosis may play an even greater role in PAD than in other arterial territories. These results also reinforce the use of lipid-lowering therapies to target disease burden across multiple vascular beds and supply the genetic rationale for targeting the coagulation cascade in PAD, as was done in VOYAGER trial2. MVP and other large databases are invaluable resources for assessing novel disease gene variants and potential genetic targets for a range of vascular diseases.

Importantly, while GWAS studies in general may be limited by confounders such as misclassification of case versus control and/or inappropriate bundling of disease subtypes under a common diagnosis, these issues may be more pronounced in PAD given the fact that many patients with disease have atypical or no symptoms, or because those with confirmed PAD are presumed to have atherosclerosis when they may in fact have some other vasculopathy. In the MVP GWAS, these issues were mitigated by chart review of a representative cohort of cases and controls to verify ICD-9 coding that correlated with clinician verified PAD diagnosis (determined through clinic notes and other diagnostic criteria). Improvements in subphenotyping with inclusion of imaging or advanced analysis of plaque characterization and disease distribution may help stratify patients in a manner reflecting distinct biology, with the potential to identify loci for specific sub-type of PAD that may be diluted by grouping all PAD patients together.

Next generation “omics” and single-cell technology:

Implementation of next-generation sequencing has become an important resource in the vascular biologist’s toolkit and may help uncover mechanisms underlying varied response to standard-of-care treatments. For example, although endarterectomy may reduce the risk of stroke in patients with symptomatic stenosis of the internal carotid artery (ICA), restenosis has been found in a substantial subset of patients, including over 35% during a three-year follow-up in one study24. This may be due to cellular heterogeneity within the plaque or the vascular wall that then influences inter-cellular behavior and disease progression. Understanding the cell types and differential gene expression patterns found in normal arterial tissue versus atherosclerotic plaques could help to refine our understanding of why some patients fail treatments such as endarterectomy or stenting. Similarly, although antiplatelet therapy is a mainstay for patients with PAD, the prevalence of antiplatelet resistance is comparable to rates seen in cardiovascular disease25, underscoring the importance of delineating cellular mechanisms leading to such variations in treatment efficacy.

Bulk sequencing studies, while yielding important overall genomic insight, obfuscate the differences between cell types contributing to gene expression. Recent efforts have focused on interrogation of the genome and its accessibility at the single-cell level. Leveraging single-cell technology to examine cellular subpopulations and genomic differences presents an important opportunity to identify disease mechanism and exploit novel targets for therapy. For example, single-cell RNA sequencing has shed critical insight into cardiac vascular differentiation26, endothelial specification27, clonal smooth muscle cell expansion28, and hematopoietic potential29. In a murine hindlimb ischemia model a small group of fibroblasts that expressed some endothelial cell genes were further dissected and clustered by scRNAseq, revealing subsets of tissue fibroblasts that seem to contribute to angiogenesis under ischemic conditions30 Many recent efforts have utilized single-cell RNA sequencing to interrogate the plaque microenvironment due to its complex cellular composition3136. We have shown transcriptional differences in carotid plaque samples between the most advanced portion of the atherosclerotic core and a region directly proximal to this location, suggesting a possible anatomic framework for atherogenesis and the possibility of site-specific disruption of disease progression36. While the majority of these studies utilize more easily obtainable tissues such as plaque from carotid endarterectomy, there is an opportunity to explore other vascular beds subjected to variable hemodynamic stressors which may shed light on site-specific genomic differences in plaque burden. For example, atherosclerotic plaque may be obtained from femoral endarterectomies for dissociation and sequencing, or alternatively full thickness artery may be harvested from lower extremity revascularization for critical limb threatening ischemia (CLTI). Transcriptome comparisons may be made between the various vascular beds sampled, leading to potentially novel and significant genomic insight into anatomic disease burden.

Refined examination of intercellular communication and cellular plasticity, both well-appreciated phenomena in vascular biology, further underscores the utility of next generation sequencing for molecular target identification. For example, vascular smooth muscle cells (VSMCs) have been shown to stimulate platelets through CLEC-2, possibly facilitating thrombus formation after plaque erosion37. Delineating which cell subtypes are responsible through single-cell analyses may allow for more targeted perturbation of this process, especially as cell-specific nanotherapies evolve38. Furthermore, the phenotypic landscape of VSMCs is understood to be quite dynamic as these cells often transition to multiple alternative phenotypes in the atherosclerotic environment39, 40. VSMCs participate in phenotype switching between contractile and synthetic subtypes, and they contribute to the development of extracellular matrix producing cells40. Advances in sequencing-based lineage tracing may facilitate mapping of clonal histories of VSMCs and other vascular cell types41, in addition to improved resolution of clonal substructure as seen in cells within the tumor microenvironemnt42. Additionally, many disease promoting processes are mediated through molecular crosstalk between VSMCs43, but this type of crosstalk is not limited to signaling between the same cell types. Atherogenesis involves coordinated genomic and molecular communication between the plethora of immune cell subtypes and other cellular components of the arterial wall44, 45. Single-cell technology offers an exciting opportunity to begin decoding this intercellular communication, while also incorporating gene network analysis which, by integration with single-cell RNA sequencing, may shed insight into the underlying biology of groups of cells36, 46.

Beyond the transcriptome, detecting chromatin accessibility with single-cell ATACseq can unveil layers of biological complexity underlying the transcriptional differences observed between cells47. Because the majority of complex disease genetic variation is non-coding48, and functional non-coding gene elements are involved in epigenetic regulation of gene expression, single-cell ATACseq has been used for chromatin-mapping to facilitate analysis of cis- and trans- regulatory elements49. Integration with single-cell RNA seq data further allows for refined cell subpopulation analysis through reconstruction of chromatin accessibility profiles of cell types defined by RNA profiles50, 51. This has enabled high-resolution examination of cardiac progenitor cell fates52, with emerging opportunity to employ this technology to assess chromatin accessibility in the peripheral vasculature.

DNA sequence variations, alternative splicing of mRNA, post-translational modifications, phosphorylation and proteolytic cleavage collectively contribute to the diversity of the human proteome far beyond the known 20,000 protein-coding genes5355. CITE-seq allows for simultaneous analysis of the transcriptome and proteome via exploitation of DNA barcodes conjugated to antibodies to enable cell surface protein detection56, 57. It has been used in conjunction with single-cell RNA sequencing for immunophenotyping of carotid plaques in order to delineate T-cell and macrophage subsets in symptomatic versus asymptomatic carotid plaques58. Emerging single-cell mass cytometry methods will allow analysis of proteins and their interactions and degradation59, bringing a unique tool for evaluation of protein signaling dynamics in the vasculature.

Single-cell analysis of the genome, epigenome, transcriptome and proteome has reshaped the ability to harness genetic variation and heterogeneity to identify and exploit novel therapeutic targets. Once identified through genomics approaches, these targets can then undergo functional analysis through in vitro and in vivo model system assays for feasibility and proof-of-concept before reaching clinical trials.

Multi-omic characterization of large, well-phenotyped clinical cohorts to discover new mechanisms and markers:

Understanding the complex interplay between traditional, environmental and host genome risk factors in determining the susceptibility to PAD requires consideration of biological networks. Tremendous advances have been made in the high throughput measurement and unbiased analysis of the “metabolome”, “proteome”, “lipidome” and the “transcriptome”10. However, to make the most of the power of these technologies, large cohorts with excellent clinical and image phenotyping are required. Application of network analytics and machine learning algorithms have the potential to uncover biological signaling pathways and mechanisms that do not rely on prior understanding. From a pragmatic perspective, greater access to tissue and atherosclerotic plaque from surgical procedures offers considerable advantages compared to similar approaches applied to the coronary or intracerebral disease process. Strategic leadership has led to the establishment of a number of large collaborative bioresources, including the Munich Vascular Biobank with biomaterial from over 1500 high-grade carotid artery stenoses; >700 PAD, and >480 aortic aneurysm cases60; and the Biobank of Karolinska Endarterectomies (BiKE) with >200 samples61. The concomitant collection of peripheral blood at the time of surgery provides the opportunity to identify a circulating biosignature of particular plaque phenotype- and particular vulnerability62. The UK Biobank provides additional opportunities based on large scale imaging (100,000 participants) of carotid plaques63.

Such approaches may also unravel some of the mysteries driving an individual’s differential susceptibility to atherosclerosis in different vascular beds (e.g. coronary versus aortic or femoral). Furthermore, the cataloging of samples based on gender and ethnicity, as well as stratification according to specific phenotypes of atherosclerosis and vascular disease (e.g. different patterns of calcification, tortuosity, morphology, histology, inflammation, neovascularization)60, may be combined with multi-omic analysis in large cohorts may provide insights to therapeutic targets relevant for specific groups.

“The formation of large biorepositories present important opportunities for precision medicine and genomics, but include challenges with infrastructure, access to comparison tissue, and sample annotation, all of which are areas of active research for improvement64.

Preclinical validation: Translational models to unravel mechanisms and test new therapies

The technologies employed to develop new drugs and products once definitive targets have been identified are rapidly expanding, and approaches such as in silico screening, fragment-based drug discovery and tagged libraries have transformed the pathways available for modern drug discovery65. However, collaborative efforts and strategic leadership are required to ensure that the mechanisms directly relevant to unmet need in human disease are considered in the choice of in vitro and in vivo models to test such compounds. This will allow the more efficient and coordinated translation of new, urgently needed therapies.

In vitro and ex vivo vascular models:

Before advancing to in vivo animal studies, investigators commonly deploy an array of pharmacological studies which rely heavily on in vitro and ex vivo cellular models. This allows for screening of chemical compound libraries and specific novel candidate drugs or products to refine and optimize putative translational targets. Because the PAD patient commonly experiences ischemia due to insufficient blood flow, a longstanding goal for the vascular research community is to develop therapies that stimulate the sprouting of new blood vessels (angiogenesis) and/or trigger the maturation of pre-existing collateral networks (arteriogenesis). There are several well-established monoculture models that have been used to specifically examine the potential beneficial effects on angiogenesis as it relates to PAD. Here we discuss both the long-standing monolayer culture approaches, as well as the more recent approaches involving 3D spheroids, co-culture, and microphysiological systems.

In vitro functional assays provide a valuable tool for assessing the effect of angiogenesis. They utilize basic cell culture techniques which can be quickly and easily performed and analyzed quantitatively. They are essential for investigating the biochemical, cellular, and molecular mechanisms occurring in angiogenesis because individual components of angiogenesis can be assessed in isolation. Endothelial cells (ECs) undergo multiple processes during angiogenesis including cell proliferation, migration, sprouting, branching and tubule formation, as well as complex cell-cell interactions. In addition, ECs differ depending on their vascular bed and vessel type – the most commonly used EC type in angiogenic assays include HUVECs, HAECs and HMECs66.

Proliferation –

The process of EC proliferation can be quantified using several different methods in vitro. The simplest method for assessing EC proliferation is counting cells. This can be achieved using the common hemocytometer and a viability dye, such as trypan blue, or through the use of automated cell counters. Colormetric assays like the MTT or the WST-1 assay are widely used to assess the natural by-product of cellular division67. Essentially these assays determine cell viability more so than proliferation68, however they are commonly used as methods to quantify EC proliferation in angiogenesis. Another potentially more accurate method used to assess proliferation includes quantifying DNA synthesis via BrdU incorporation69 or Click-iT via EdU incorporation70.

Migration –

After proliferation, ECs migrate away from their origin. All angiogenic models involve the sprouting of ECs from a monolayer, and the movement of ECs through the basement membrane is often referred to as “sprouting” and occurs due to nearby chemical signals that either promote or prevent angiogenic activity. Assessing the migratory ability of ECs can be achieved with a number of different assays each with their own advantages and disadvantages. Wound healing or the ‘scratch’ assay is the simplest and quickest assay to perform. ECs grown to a confluent monolayer are ‘wounded’ commonly with a pipet tip or other similar hard objects to create a denuded zone71. In response to this damage, ECs begin to proliferate and migrate into the denuded zone in order to heal the wound. The clear disadvantage of this assay is the reproducibility with issues in confluency of cells, consistency of the scratch and difficulty in accurate quantitation71. Transfilter assays or Boyden chamber assays are generally used to study migration of ECs towards a stimulus (commonly VEGF) and have the advantage of distinguishing between specific directional migration instead of random cell movement69. ECs are seeded into the upper chamber with the stimulus in the lower chamber in culture medium. The migration of ECs through the filter can take anywhere from several hours to 48 hours and can be quantified by cell staining or counting of cells attached to the filter. The main weakness of this assay is the inability to observe real-time migration. New automated technologies are now able to overcome this limitation with the use of systems such as xCELLigence (Roche)72 and also the electric cell-substrate impedance sensing (ECIS) instrument (Applied Biophysics)73. These systems allow real-time monitoring of cell proliferation and migration when cells are grown in special electrode covered chambers. The resistance generated by the cell membrane increases the electrical potential between the electrodes and this allows monitoring of the cell behavior66.

Tube Formation –

Tubulogenesis, more commonly termed tube formation, is the connection of the new blood vessels to preexisting vessels beds. Modelling this process in vitro is a cornerstone of angiogenic research. Using ECs grown in culture and then placed on commercially available Matrigel®, this assay is a two-dimensional imitation of extracellular matrix. The Matrigel acts as the extracellular matrix containing cytokines and growth factors allowing the ECs to form tubule-like structures67. Tube formation can be observed with imaging techniques and can take anywhere from 3–24 hours to provide reliable data. The results obtained can be highly variable and comparisons in the literature depend widely on cell type, quantity of Matrigel used, and endpoint analyzed (tubule number, tubule length or branch points)69. Furthermore, different methods of analysis allow for increased variability, whether these data points are measured manually, which is more arduous and time-consuming, or using automated image analysis programs, which require screening to eliminate artifacts but can quantify at a faster rate. This 2D model has since been expanded to 3D assays to more closely mimic the angiogenic sprouting of ECs observed in vivo69. Simple conversions of this assay include using thicker basement matrix so ECs can be seeded at differing levels to observe tubule formation both vertically and horizontally. More complex versions include scaffold-free cell sheet stacking, cell aggregate microtissue assays and microcarrier spherical scaffold assays66, 74. All of these have the advantage of recapitulating all the stages of in vivo angiogenesis however the technology required for visualization and analysis can be costly and difficult to interpret. 3D models have also expanded to include co-culture- incorporating not just endothelial cells, but also vascular smooth muscle cells and fibroblasts19, 75. Such approaches may enhance the modelling of in vivo human disease, and have the capacity to support early drug screening, potentially reducing the number of animal studies required. However, it is essential we improve the uniformity of organoid models at the necessary scale. Proof of translatability is still a major issue, similar to all preclinical models in the PAD space.

Embryoid body vasculogenesis assay –

Embryonic stem cells (ES cells) have been highlighted as promising cell sources for the study of differentiation towards the endothelial lineage and vasculogenesis7678 since they can go through the majority stages of angiogenesis observed in vivo79. This assay requires ES cells to be initially differentiated into embryoid bodies (EBs) which are then placed into a 3D collagen matrix for the EB primary vascular structures to extend and invade the collagen matrix, leading to sprouting of ECs similar to the angiogenic process80. Using cell imaging, this assay allows for qualitative and quantitative analysis of multiple steps in angiogenesis and makes it possible to test many different therapeutic agents81. While the assay is comprehensive it is time consuming both at the experimental and analytical stages82. Additionally, the use of ES cells in biomedical research has been hindered by ethical issues since extraction of these cells disrupts the human embryo83, 84. Even with several studies demonstrating effective use of these cells in the study of angiogenesis, ES cells are not generally considered an ethical source of endothelial cells80.

iPSCs – “disease in a dish” –

Overcoming the ethical issues with the use of ES cells, a breakthrough discovery in cellular biology was the discovery of induced pluripotent stem cells (iPSC) made by Takahashi in 200685. With advances in cellular reprogramming, iPSCs are derived from adult somatic cells86 and blood87, 88, into a stem-cell like state by the introduction of the common transcription factors OCT4, SOX2, KLF4 and c-MYC (Yamanaka factors) via viral transduction, protein and microRNA transduction, or by chemical/small molecule-based reprogramming strategies89. The first use of iPSC in the field of vascular biology occurred in 2009, when Taura et al. demonstrated the ability to yield endothelial cells from iPSC (iPSC-EC), which when matured display phenotypic properties highly similar to primary endothelial cells90, 91. More recently, iPSC-ECs have shown therapeutic potential by enhancing angiogenesis in mouse models with improvements in blood reperfusion and enhanced wound closure92, 93. A major advantage of iPSCs is that they can be easily generated from patients with or without disease for use in angiogenic assays94. Blood vessel development can be modelled in 3D in vitro assays using iPSC-ECs cultured within engineered platforms that mimic the 3D microenvironment95. Hydrogel scaffolds, either on standard well plates or within a passive pumping microfluidic device have offered a more defined platform for vascular-based assays96, 97. Major advances in genome editing techniques with the use of clustered regularly interspaced palindromic repeats/Cas9 (CRISPR/Cas9) allows causality of genetic variants to be explored in a rigorous manner in patient-derived iPSCs and derived vascular cells98 with full characterization of cell signaling and molecular profiling. As the application of iPSC-derived vascular cells and models evolve, it will be critical for the field to develop standardized protocols86, 99.

Aortic Ring Assays –

Beyond the standard assays outlined above, investigators commonly employ organ explant assays, which appear to synergistically assess almost all steps of physiological angiogenesis100. Aortic ring assays, often termed “ex vivo” models, have been developed to be the in vitro mimic of in vivo angiogenesis with the ability to measure angiogenic sprouting, outward growth, and (to an extent) stabilization of new blood vessels from explanted segments of vasculature101, 102. Vessel rings are excised, cleaned, cut into ~1mm sections, immersed in collagen, fibrin or Matrigel and cultured for vessel outgrowth. After a few days, the outgrowth can be measured using staining and microscopy, then compared to samples that have been exposed to potential angiogenic agents. The advantages of the aortic ring assay are numerous including the capture of multiple cell organization with additional paracrine support and extracellular matrix components69, 100. The downside of this assay is reproducibility due to imprecise segments and the absence of blood flow effects.

Vascular organoids (scaffold free) –

The assembly of different cell types into 3D spheroid co-cultures called ‘vascular organoids’ are being described as a bridge in the gap between traditional 2D in vitro cell culture and the complexity of animal models. These transformational 3D model systems provide a more physiologically relevant assay encompassing complex cell-cell interactions which enables the deposition of their own extracellular matrix103, 104. The advent of hybrid spheroids using more than one cell type provide useful models for angiogenesis assays with the formation of tubular-like structures105, 106. Multiple different techniques including the hanging-drop, the use of low adhesion plates or self-organising vessel-like structures have been utilized to assess angiogenesis. While these methods can be relatively cost-effective they are time-consuming and require advanced imaging capabilities for analysis107. More recently, these methods utilize the addition of iPSC-ECs with multiple other cell types in order to increase the ability for cells to reorganise and form complex vascularized networks108, 109. Studies have shown that after implantation of iPSC-EC organoids into the chick chorioallantoic membrane assay, the generated vessels connect with the host circulation and vessel structure is preserved109. Many aspects of vessel formation within the vascular organoid can be detected including branching, an extracellular matrix, cell-cell interaction, microvesicle release and responsiveness to pro- and antiangiogenic stimuli110.

Vascular scaffolds –

In addition to the more commonly used scaffold of Matrigel to assess EC interactions in angiogenic-based assays, other more complex vascular networks are being bioengineered to incorporate the missing in vivo elements. Electrospinning and 3D bioprinting are being utilized to address the need for a more complete microenvironment including cell organization, spatial control and localized release of growth factors in response to cues from the extracellular matrix and localized blood flow. Electrospinning can produce nanofiber-based synthetic vascular networks with similar dimensions to naturally occurring extracellular matrix (50–500nm) and provides an excellent platform for the adhesion of ECs in a monolayer. A limitation in this methodology however, is that nanofibers create a physical barrier for cells and this limits the ability to mimic the permeability of the endothelial monolayer111. 3D bioprinting is an emerging approach to generate scaffolds with hydrogel which can be embedded with microchannels to study angiogenesis112, 113. Using either direct or indirect printing methods, cells, biomaterials, and growth factors can be combined to produce complex shaped constructs with defined micron-sized channels and pore sizes that are capable of guiding angiogenesis. Both electrospinning and 3D printing techniques have various advantages and are frequently used in tissue engineering applications. The 3D bioprinting technique allows controlling the production of a large number of scaffolds with precise measurements relatively quickly while electrospinning builds scaffolds with a wide range of properties to mimic blood vessel structure including composition, diameter, thickness, porosity, and degradation rates113.

Nanoparticles –

The testing of targeted angiogenic pathways has been significantly enhanced by technological advances in nanotechnology. More efficacious administration of angiogenesis modulators using nanoparticles, including metallic, metal oxide, glass-ceramic and polymeric nanoparticles, has allowed for treatments with a longer half-life and more selective targeting of the vasculature114117. Some of the first studies used albumin and gelatin nanoparticles containing a DNA plasmid to trap excess VEGF and reduce angiogenesis118, while alternative applications were designed to increase angiogenesis and enhance revascularization118120. Clinical trials involving VEGF therapy have been unsuccessful at improving peak walking time, ankle-brachial index, or quality of life121, 122, thus underscoring the need to evaluate each step of the translational cascade described.

Other platforms include single- and multi-walled nanotubes containing micro-RNA oligonucleotides and VEGF-targeted siRNA, able to regulate EC proliferation and prevent angiogenesis123, 124. Graphene oxide nanoparticles structures have demonstrated increased proliferation and migration of cells to promote wound healing and nano diamonds can increase bioavailability of angiogenic drugs for improved delivery to tissue125, 126. Among metallic options, gold nanoparticles are being recognized for their angiogenic potential117. They are composed of an inorganic core containing gold encircled by an organic monolayer and have displayed numerous anti-angiogenic effects by suppressing activation of VEGFR2, Tie2R, FGFR and many of their downstream signaling pathways114, 127, 128. While the majority of these applications have been targeted for tumor-related angiogenesis and therefore focus on anti-angiogenic properties, there are some applications that have demonstrated pro-angiogenic effects129, with increased cell survival and proliferation of ECs and increased vessel-like structures, particularly in wound healing130, 131. Nanoparticle characterization is increasingly variable and therefore difficult to compare across multiple studies. Given the missing details in some publications there has been a call for “minimum information reporting” with regards to the details in the study of nanomaterials and their biological interactions132. Obtaining a standard in the field will improve the growth of available and effective nanoparticles for use in angiogenesis.

Specific vascular targeting nanoparticles continue to be developed in an effort to advance precision therapy for atherosclerosis. Lesional macrophage-specific single walled nanotubes (SWNTs) are shown to accumulate in the atherosclerotic plaque and, with the appropriately coupled therapeutic, reactivate efferocytosis in order to reduce plaque burden with minimal off-target cytotoxicity38. Targeting of vascular inflammation using leukosomes, a biomimetic nanoparticle that integrates leukocyte-derived membrane proteins into the phospholipid bilayer, has been shown to enhance delivery of therapeutic to inflamed endothelium at sites of plaque development133. Continued efforts to refine cell-type specific therapeutic delivery will likely be critical for future vascular therapies.

Pre-clinical in vivo models:

Once a novel target gene or therapeutic candidate has been identified, the preceding in vitro assays are commonly used to determine which leads should be prioritized for additional in vivo testing. In the field of vascular medicine, investigators tend to be broadly focused on determining whether a given pathway may regulate new blood vessel formation, peripheral atherosclerosis, ischemic tissue wound healing, and/or thrombosis134. In the following section, we provide a general overview of the in vivo mouse models available to test these processes, including the strengths and limitations of each approach, as well as emerging techniques that may allow more sophisticated analyses of the biology underlying peripheral vascular disease.

Hind limb ischemia models:

As discussed previously, mitigation of ischemic complications in PAD through stimulation of angiogenesis and arteriogenesis are longstanding goals in vascular research. These processes can be assessed several ways (including with retinal angiogenesis or Matrigel implantation assays) but are most commonly tested in the murine hindlimb ischemia (HLI) model135, 136. This model relies on the interruption of lower extremity blood flow, most often induced by the ligation of the femoral artery (which can model claudication given that collaterals downstream of the profunda femoral artery are left intact) or via the simultaneous ligation and excision of the femoral artery (which can model critical limb ischemia given that the collateral circulation is also perturbed with this approach). Limb perfusion is then quantified by noninvasive laser Doppler imaging, where the flux of blood (normalized to the non-ischemic limb) can be quantified during recovery or in response to therapeutic intervention. Other reproducible readouts include the use of microCT angiography, digital necrosis scoring, and blinded histological quantification of neovessel density19. Studies in large animal models of PAD have had success in mimicking the ischemic myopathy seen in ischemic human limbs by using endovascular techniques and occluding inline flow through the external iliac artery and impeding the rapid arterial collateralization through the use of a covered stent137.

While the HLI model is commonly used, it does have several limitations that warrant discussion136. First, these studies are most often performed in young, non-atherosclerotic mice without any of the comorbidities typically encountered in humans with PAD. While some have advocated for the use of diabetic (e.g. db/db) or dyslipidemic (e.g. apoE−/−) animals, it is important to note that the acute cessation of flow in an otherwise healthy artery is perhaps more reflective of acute limb ischemia (ALI) than the gradual reduction in perfusion that is observed in vasculopaths who experience reductions in their ankle brachial indices (ABI) over a period of months to years. Models that utilize an ameroid constrictor device (which can gradually occlude the vessel) have been reported and are being studied as a potentially more relevant simulator of human vascular disease138. Second, it is critical to recognize that there are profound differences across mouse strains which can influence the interpretation of results with this model139. For example, the widely used C57Bl/6 strain has robust collateral reserve and is resistant to critical limb ischemia, while the BALB/c strain is highly susceptible to digital necrosis and even limb loss after femoral ligation. Thus, investigators must consider whether they are interested in modelling claudication or CLI before generating a new genetic knockout model or embarking on a therapeutic intervention study. While multiple modalities can be used to quantify limb perfusion, each has its own intrinsic limitations, as previously summarized by Lotfi and colleagues136. In the particular case of laser Doppler imaging, factors that can alter vasoconstriction or vasomotor tone (e.g. depth of sedation, room temperature, animal sex, etc.) should be controlled to the best extent possible140. Finally, studies exploring the cell-cell interactions occurring during limb perfusion are providing mechanistic insights into vascular repair. More than simply an EC proliferative response, the involvement of VSMCs, macrophages and pericytes are contributing at a molecular level to participate in tissue perfusion.

Peripheral plaque vulnerability models:

Given that most PAD is atherothrombotic in nature141, the translational vascular research community also employs a wide array of mouse models that develop atherosclerotic plaques in the peripheral vasculature, including the aorta and brachiocephalic artery. Traditional dyslipidemia models (e.g. LDLR−/− and apoE−/− mice fed a high fat ‘Western’ diet) can be used to reproducibly quantify plaque burden (using lipid stains such as Oil Red O) and lesion composition (with histological analysis of necrotic core size, foam cell burden, and fibrous cap thickness). However, most commonly used murine atherosclerosis models are not susceptible to spontaneous plaque rupture, and do not consistently demonstrate the intraplaque hemorrhage and superimposed luminal thrombosis encountered in subjects with PAD. Accordingly, a variety of models that induce mechanical alterations in blood flow to promote endothelial dysfunction and plaque-destabilization have now been described. These methods (which include interposition grafting, perivascular device placement, arterial ligations, or fistula formation) were recently reviewed by Winkel and colleagues142, including their predicted impact on shear stress and how those changes can be used to model various aspects of peripheral vascular disease143.

Amongst these models, the application of an extrinsic silastic collar to the carotid artery (with144 or without145, 146 angiotensin infusion) seems to have emerged as the approach which most faithfully recapitulates the human condition147. While technical variations in the approach exist148, these so-called ‘cuff’ or ‘cast’ models promote oscillatory and/or reduced shear stress in the branch vessels of the aortic arch, thus leading to the generation of advanced and destabilized carotid lesions. These plaques demonstrate a high burden of macrophage infiltrates, a reduction in collagen and smooth muscle cell (SMC) content, thinning of the fibrous cap, and intraplaque hemorrhage148. Such vulnerability models can be deployed in mice with indelible cell-specific lineage tracers, which are recognized as being highly informative given the increasingly appreciated role for phenotype switching, cellular plasticity, and clonal expansion during atherogenesis149151. Moreover, our group recently demonstrated that cast-induced changes in lesion vulnerability can be quantified non-invasively (using 18F-FDG-PET CT scanning), and that this imaging modality is sensitive enough to detect changes in plaque stability after drug treatment over time152. Accordingly, these peripheral plaque rupture models may prove useful for investigators studying new translational therapies.

Ischemic wound healing models:

Due to poor circulation and prolonged tissue ischemia, many patients with PAD develop chronic wounds or ulcers on their lower limbs. These can be very difficult to heal and often become infected and require amputation of the affected foot or leg153, 154. In addition to the health burden, the economic consequences are substantial. The complications associated with infection and amputation also lead to increased mortality in these patients, particularly in diabetic patients153. Wound healing is a process not able to be replicated in cell models and has primarily been limited to large animal models such as swine, to ensure a comparable process to humans155. In the last two decades however, murine in vivo wound healing experiments have become effective models for studying human wound healing156. Three reproducible murine wound healing models that recapitulate the human wound healing process have been described – the splinted excisional wound157, ischemia-reperfusion model158 and the ischemic flap model156. Excisional wounds on the dorsal surface are the most commonly used wound healing model, however in mice wound contraction is a significant limitation in assessing wound closure. The use of splints therefore increases the relevancy of this model to human wound healing159. Silicone splints are fixed to the wounds to prevent contraction and allow the wound to heal through tissue formation and reepithelialization155, 157. Despite this, it still remains a great challenge to replicate wound healing as murine models present different anatomy to humans, particularly for the use of rodent skin. In a recent systematic review160, animal models of ischemic wound healing were investigated with only three studies utilizing more clinically relevant wounds created on the paws of mice after undergoing hindlimb ischemia161163. Some recommendations to help improve this model includes the use of aged mice to mimic human diseases, improving bias and standardizing assessments for the ideal ischemic ulcer model in addition to using a more stable induction of ischemia138.

From bench to bedside: Accelerating translation through early phase clinical trials

While there have been substantial improvements in preclinical models of PAD, the disease complexity has contributed to disappointing results in the clinical translation of therapies showing potential at earlier stages. Collaboration at the interface of the preclinical and clinical efforts is required to improve the prioritization of the most promising novel therapies and to strategize how to overcome developmental hurdles. More efficient early phase studies focused on biologically relevant endpoints in more precisely stratified patients will be key. In addition to traditional and novel systemically administered therapies, the ‘peripheral’ and accessible nature of PAD should also make innovative approaches to local delivery of novel therapies feasible. This may involve stents or bioresorbable scaffolds, or topical nanoparticle delivery, for example to ischemic ulcers. Outlined below are concepts that may be useful in accelerating the translation of discoveries from bench to bedside.

Precision stratification of patients based on biology:

There remains a considerable degree of disease heterogeneity in PAD that directly relates to underlying biological mechanisms, as outlined above. A key consequence of such heterogeneity is dilution of the average therapeutic effect in trials, thus necessitating larger, longer and more expensive clinical development programs. Approaches described in this review, including genomics, molecular phenotyping and improved imaging techniques will provide new risk stratification tools with better opportunity to ‘divide by biology’. While pursuing broadly generalizable therapies with that can be used by all vascular patients obviously remains a key goal, testing new treatments in the groups where the therapy makes the most biological ‘sense’ may help reduce the size of studies and facilitate the demonstration of the impact of the intervention.

Identification of those with greatest unmet need:

A current unmet need in the management of cardiovascular disease is the ability to address the substantial number of individuals who develop disease or progress despite best practice treatment. While there is an obvious group of patients who develop disease due to a history of smoking and/or mixed adherence to recommended treatments, there are also many patients whose disease is driven by unknown biological susceptibility. While at a community and policy level it makes sense to invest in the widespread use of cost-effective and evidence-based care, recognizing that there are subgroups of individuals who demonstrate rapid disease progression, similar to cancer patients with metastasis on therapy, is key. Emerging machine learning and neural networking approaches are beginning to allow for highly accurate sub-phenotyping of those most susceptible to disease, or those least likely to respond to conventional therapy164. These clinical and genetic enrichment-based approaches will enhance discovery of novel biological mechanisms that are not addressed by existing agents, as well as improve our ability to rapidly test new therapies in early phase trials due to higher event rates, and smaller sample size requirements.

Enhancing our focus on biologically relevant intermediate endpoints:

Efficient translational pipelines require consideration of staged regulatory approaches. Vascular research has set itself an extremely high bar, often using Major Adverse Cardiovascular Events (MACE) or Major Adverse Limb Events (MALE) as the primary endpoints in late-stage clinical trials. Using traditional study design, and minimally stratified recruitment approaches, this high bar drives the need for massive recruitment requirements and high study costs. As a result, many important trials are slower than expected, fail to fully enroll, or run out of funding while waiting for events to accrue165. Working with regulatory authorities to derive and agree upon biologically meaningful intermediate endpoints with a direct role in disease progression and/or patient reported outcomes is therefore of critical importance. In the case of oncology, tumor size and its impact on quality of life are relatively obvious measures. In the case of PAD, consideration of vascular function, tissue perfusion, lesion “vulnerability” or degree of plaque inflammation may all be considered. In addition, rigorous definition and consensus around tissue specific endpoints may be a unique opportunity for PAD within the Cardiovascular field. This relates to our ability to access relevant tissue much more easily than cardiologists- such as at the time of carotid endarterectomy, debridement or amputation. The field could consider more strategic ways of using this in design of clinical trials- unlocking the possibility of novel biologically relevant intermediate endpoints for new therapies. Further opportunities for biologically relevant endpoints with potential for mechanistic insights include measures of vascular function (e.g. EndoPAT166) or vascular inflammation and plaque burden (FDG/PET CT167). Remembering our goal of not only helping patients live longer but also feel better, disease-specific endpoints of relevance to the PAD patient should also be more actively integrated into clinical development programs, including improvements in claudication symptoms or the ability to walk more freely at home (quantified via mobile health devices).

Future Directions:

Vascular biology research continues to build upon important discoveries by embracing the translational approaches described in this review. Next generation genomics should not be considered an adjunct, but rather a critical component of vascular studies used to elucidate mechanisms of disease at the cell-specific level. Delineation of disease process in immune cells versus cells of the vascular wall, for example, may inform future therapeutic direction. Correlation and integration of this data with population-wide GWAS studies may form the foundation upon which novel therapies are pursued in clinical trials, which clearly can be enhanced and streamlined. It is also critical to utilize and improve upon existing animal models of disease. Combining animal models, sophisticated in vitro techniques, and advanced genomics will ultimately bring important discoveries in vascular research closer to translational potential. Consideration and consensus on the use of new biologically meaningful intermediate endpoints in PAD disease progression will be crucial for future clinical trials. Finally, as investigators continue to make discoveries, new challenges emerge about how discovery turns into useful therapy for clinical practice. One consideration is that while the vasculature serves as a conduit for systemic therapeutic delivery, how are blood vessels themselves targeted in a specific and meaningful manner? Based on the disease process being studied, investigators should continue to assess whether targeting disease systemically or at its nidus would provide the most benefit. Along these lines, refinement of tissue and/or cell-specific therapeutic delivery systems will impact vascular research that relies on a targeted approach for disease amelioration. Collectively, the challenges and opportunities outlined in this review ensure an exciting future for translational vascular biology, hopefully bringing enhanced vascular care closer to patients who increasingly need it.

Figure 1.

Figure 1.

Schematic demonstrating existing and evolving state-of-the-art components to enhance bench to bedside translation in vascular research.

Figure 2.

Figure 2.

Opportunities for novel target identification and prioritization in large, well-phenotyped clinical cohorts integrating molecular and histological characterization with GWAS and single cell multi-“omic” technologies.

Figure 3.

Figure 3.

Schematic summary of preclinical cellular and animal models relevant to PAD. In vitro models include angiogenesis assays, and patient-derived co-cultured organoids with the opportunity for CRISPR gene editing. In vivo models include hind limb ischemia and wound-healing in mice and abdominal aneurysm model established in pig models.

Figure 4.

Figure 4.

Schematic clinical trial concept with example intermediate biologically and clinically relevant endpoints.

Acknowledgments

Funding Sources: GAF is supported by a National Health and Medical Research Council Practitioner Fellowship (grant number APP11359290), Heart Research Australia, and the New South Wales Office of Health and Medical Research. This review was also supported by NIH R35 HL144475 awarded to NJL, and the Ansell Fellowship awarded to TA.

Conflicts: GAF reports personal consulting fees from CSL and grants from Abbott Diagnostic outside the submitted work. In addition, GF has a patent Biomarkers and Oxidative Stress awarded USA May 2017 (US9638699B2) issued to Northern Sydney Local Health District. NJL reports personal consulting fees from Janssen.

Non-standard Abbreviations and Acronyms:

PAD

Peripheral arterial disease

GWAS

Genome wide association studies

VSMC

Vascular smooth muscle cell

EC

Endothelial cell

ATAC-seq

Assay for transposase-accessible chromatin-sequencing

CITE-seq

Cellular indexing of transcriptomes and epitopes by sequencing

iPSC

induced pluripotent stem cell

SWNT

Single walled nanotube

MACE

Major adverse cardiac events

MALE

Major adverse limb events

References:

  • 1.Kullo IJ and Rooke TW. Peripheral Artery Disease. New England Journal of Medicine. 2016;374:861–871. [DOI] [PubMed] [Google Scholar]
  • 2.Bonaca MP, Bauersachs RM, Anand SS, Debus ES, Nehler MR, Patel MR, Fanelli F, Capell WH, Diao L, Jaeger N, et al. Rivaroxaban in Peripheral Artery Disease after Revascularization. N Engl J Med. 2020;382:1994–2004. [DOI] [PubMed] [Google Scholar]
  • 3.Murphy TP, Hirsch AT, Ricotta JJ, Cutlip DE, Mohler E, Regensteiner JG, Comerota AJ, Cohen DJ and Committee CS. The Claudication: Exercise Vs. Endoluminal Revascularization (CLEVER) study: rationale and methods. Journal of vascular surgery. 2008;47:1356–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kullo IJ and Rooke TW. CLINICAL PRACTICE. Peripheral Artery Disease. N Engl J Med. 2016;374:861–71. [DOI] [PubMed] [Google Scholar]
  • 5.Weber C and Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–22. [DOI] [PubMed] [Google Scholar]
  • 6.Aday AW and Everett BM. Dyslipidemia Profiles in Patients with Peripheral Artery Disease. Curr Cardiol Rep. 2019;21:42–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Safar ME, Priollet P, Luizy F, Mourad JJ, Cacoub P, Levesque H, Benelbaz J, Michon P, Herrmann MA and Blacher J. Peripheral arterial disease and isolated systolic hypertension: the ATTEST study. J Hum Hypertens. 2009;23:182–7. [DOI] [PubMed] [Google Scholar]
  • 8.Matsagas MI, Geroulakos G and Mikhailidis DP. The role of platelets in peripheral arterial disease: therapeutic implications. Ann Vasc Surg. 2002;16:246–58. [DOI] [PubMed] [Google Scholar]
  • 9.Kott KA, Vernon ST, Hansen T, Yu C, Bubb KJ, Coffey S, Sullivan D, Yang J, O’Sullivan J, Chow C, et al. Biobanking for discovery of novel cardiovascular biomarkers using imaging-quantified disease burden: protocol for the longitudinal, prospective, BioHEART-CT cohort study. BMJ Open. 2019;9:e028649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Vernon ST, Hansen T, Kott KA, Yang JY, O’Sullivan JF and Figtree GA. Utilizing state-of-the-art “omics” technology and bioinformatics to identify new biological mechanisms and biomarkers for coronary artery disease. Microcirculation. 2019;26:e12488. [DOI] [PubMed] [Google Scholar]
  • 11.Cohen JC, Boerwinkle E, Mosley TH Jr. and Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354:1264–72. [DOI] [PubMed] [Google Scholar]
  • 12.Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK and Hobbs HH. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37:161–5. [DOI] [PubMed] [Google Scholar]
  • 13.Abifadel M, Varret M, Rabes JP, Allard D, Ouguerram K, Devillers M, Cruaud C, Benjannet S, Wickham L, Erlich D, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34:154–6. [DOI] [PubMed] [Google Scholar]
  • 14.Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, et al. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation. 2019;139:e56–e528. [DOI] [PubMed] [Google Scholar]
  • 15.Libby P, Bornfeldt KE and Tall AR. Atherosclerosis: Successes, Surprises, and Future Challenges. Circ Res. 2016;118:531–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nikpay M, Goel A, Won H-H, Hall LM, Willenborg C, Kanoni S, Saleheen D, Kyriakou T, Nelson CP, Hopewell JC, et al. A comprehensive 1000 Genomes–based genome-wide association meta-analysis of coronary artery disease. Nature Genetics. 2015;47:1121–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Samani NJ, Erdmann J, Hall AS, Hengstenberg C, Mangino M, Mayer B, Dixon RJ, Meitinger T, Braund P, Wichmann HE, et al. Genomewide association analysis of coronary artery disease. N Engl J Med. 2007;357:443–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cluett C, McDermott MM, Guralnik J, Ferrucci L, Bandinelli S, Miljkovic I, Zmuda JM, Li R, Tranah G, Harris T, et al. The 9p21 myocardial infarction risk allele increases risk of peripheral artery disease in older people. Circ Cardiovasc Genet. 2009;2:347–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nanda V, Downing KP, Ye J, Xiao S, Kojima Y, Spin JM, DiRenzo D, Nead KT, Connolly AJ, Dandona S, et al. CDKN2B Regulates TGFβ Signaling and Smooth Muscle Cell Investment of Hypoxic Neovessels. Circ Res. 2016;118:230–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gaziano JM, Concato J, Brophy M, Fiore L, Pyarajan S, Breeling J, Whitbourne S, Deen J, Shannon C, Humphries D, et al. Million Veteran Program: A mega-biobank to study genetic influences on health and disease. Journal of Clinical Epidemiology. 2016;70:214–223. [DOI] [PubMed] [Google Scholar]
  • 21.Klarin D, Lynch J, Aragam K, Chaffin M, Assimes TL, Huang J, Lee KM, Shao Q, Huffman JE, Natarajan P, et al. Genome-wide association study of peripheral artery disease in the Million Veteran Program. Nature Medicine. 2019;25:1274–1279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Brown DW. Smoking prevalence among US veterans. J Gen Intern Med. 2010;25:147–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Selim AJ, Berlowitz DR, Fincke G, Cong Z, Rogers W, Haffer SC, Ren XS, Lee A, Qian SX, Miller DR, et al. The health status of elderly veteran enrollees in the Veterans Health Administration. J Am Geriatr Soc. 2004;52:1271–6. [DOI] [PubMed] [Google Scholar]
  • 24.Oszkinis G, Pukacki F, Juszkat R, Weigele JB, Gabriel M, Krasinski Z, Zielinski M and Krejza J. Restenosis after carotid endarterectomy: incidence and endovascular management. Interv Neuroradiol. 2007;13:345–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Guirgis M, Thompson P and Jansen S. Review of aspirin and clopidogrel resistance in peripheral arterial disease. J Vasc Surg. 2017;66:1576–1586. [DOI] [PubMed] [Google Scholar]
  • 26.Su T, Stanley G, Sinha R, D’Amato G, Das S, Rhee S, Chang AH, Poduri A, Raftrey B, Dinh TT, et al. Single-cell analysis of early progenitor cells that build coronary arteries. Nature. 2018;559:356–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Paik DT, Tian L, Lee J, Sayed N, Chen IY, Rhee S, Rhee JW, Kim Y, Wirka RC, Buikema JW, et al. Large-Scale Single-Cell RNA-Seq Reveals Molecular Signatures of Heterogeneous Populations of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Circ Res. 2018;123:443–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wang Y, Nanda V, Direnzo D, Ye J, Xiao S, Kojima Y, Howe KL, Jarr K-U, Flores AM, Tsantilas P, et al. Clonally expanding smooth muscle cells promote atherosclerosis by escaping efferocytosis and activating the complement cascade. Proceedings of the National Academy of Sciences. 2020;117:15818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Baron CS, Kester L, Klaus A, Boisset JC, Thambyrajah R, Yvernogeau L, Kouskoff V, Lacaud G, van Oudenaarden A and Robin C. Single-cell transcriptomics reveal the dynamic of haematopoietic stem cell production in the aorta. Nat Commun. 2018;9:2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Meng S, Lv J, Chanda PK, Owusu I, Chen K and Cooke JP. Reservoir of Fibroblasts Promotes Recovery From Limb Ischemia. Circulation. 2020;142:1647–1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rekhter MD and Gordon D. Active proliferation of different cell types, including lymphocytes, in human atherosclerotic plaques. Am J Pathol. 1995;147:668–77. [PMC free article] [PubMed] [Google Scholar]
  • 32.Winkels H, Ehinger E, Vassallo M, Buscher K, Dinh HQ, Kobiyama K, Hamers AAJ, Cochain C, Vafadarnejad E, Saliba AE, et al. Atlas of the Immune Cell Repertoire in Mouse Atherosclerosis Defined by Single-Cell RNA-Sequencing and Mass Cytometry. Circ Res. 2018;122:1675–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Winkels H, Ehinger E, Ghosheh Y, Wolf D and Ley K. Atherosclerosis in the single-cell era. Curr Opin Lipidol. 2018;29:389–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Depuydt MA, Prange KH, Slenders L, Örd T, Elbersen D, Boltjes A, de Jager SC, Asselbergs FW, de Borst GJ, Aavik E, et al. Microanatomy of the Human Atherosclerotic Plaque by Single-Cell Transcriptomics. Circ Res. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cochain C, Vafadarnejad E, Arampatzi P, Pelisek J, Winkels H, Ley K, Wolf D, Saliba AE and Zernecke A. Single-Cell RNA-Seq Reveals the Transcriptional Landscape and Heterogeneity of Aortic Macrophages in Murine Atherosclerosis. Circ Res. 2018;122:1661–1674. [DOI] [PubMed] [Google Scholar]
  • 36.Alsaigh T, Evans D, Frankel D and Torkamani A. Decoding the transcriptome of atherosclerotic plaque at single-cell resolution. bioRxiv. 2020:2020.03.03.968123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Inoue O, Hokamura K, Shirai T, Osada M, Tsukiji N, Hatakeyama K, Umemura K, Asada Y, Suzuki-Inoue K and Ozaki Y. Vascular Smooth Muscle Cells Stimulate Platelets and Facilitate Thrombus Formation through Platelet CLEC-2: Implications in Atherothrombosis. PLoS One. 2015;10:e0139357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Flores AM, Hosseini-Nassab N, Jarr K-U, Ye J, Zhu X, Wirka R, Koh AL, Tsantilas P, Wang Y, Nanda V, et al. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat Nanotechnol. 2020;15:154–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liu M and Gomez D. Smooth Muscle Cell Phenotypic Diversity. Arterioscler Thromb Vasc Biol. 2019;39:1715–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR and Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nature Reviews Cardiology. 2019;16:727–744. [DOI] [PubMed] [Google Scholar]
  • 41.Wagner DE and Klein AM. Lineage tracing meets single-cell omics: opportunities and challenges. Nature Reviews Genetics. 2020;21:410–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gao R, Bai S, Henderson YC, Lin Y, Schalck A, Yan Y, Kumar T, Hu M, Sei E, Davis A, et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nature Biotechnology. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chen PY, Qin L, Li G, Tellides G and Simons M. Smooth muscle FGF/TGFbeta cross talk regulates atherosclerosis progression. EMBO Mol Med. 2016;8:712–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Libby P, Lichtman AH and Hansson GK. Immune effector mechanisms implicated in atherosclerosis: from mice to humans. Immunity. 2013;38:1092–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hu D, Yin C, Luo S, Habenicht AJR and Mohanta SK. Vascular Smooth Muscle Cells Contribute to Atherosclerosis Immunity. Frontiers in Immunology. 2019;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jackson CA, Castro DM, Saldi GA, Bonneau R and Gresham D. Gene regulatory network reconstruction using single-cell RNA sequencing of barcoded genotypes in diverse environments. Elife. 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, Chang HY and Greenleaf WJ. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature. 2015;523:486–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS and Manolio TA. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proceedings of the National Academy of Sciences. 2009;106:9362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Baek S and Lee I. Single-cell ATAC sequencing analysis: From data preprocessing to hypothesis generation. Computational and Structural Biotechnology Journal. 2020;18:1429–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cao J, Cusanovich DA, Ramani V, Aghamirzaie D, Pliner HA, Hill AJ, Daza RM, McFaline-Figueroa JL, Packer JS, Christiansen L, et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science (New York, NY). 2018;361:1380–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Paik DT, Cho S, Tian L, Chang HY and Wu JC. Single-cell RNA sequencing in cardiovascular development, disease and medicine. Nature Reviews Cardiology. 2020;17:457–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jia G, Preussner J, Chen X, Guenther S, Yuan X, Yekelchyk M, Kuenne C, Looso M, Zhou Y, Teichmann S, et al. Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement. Nature Communications. 2018;9:4877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Labib M and Kelley SO. Single-cell analysis targeting the proteome. Nature Reviews Chemistry. 2020;4:143–158. [DOI] [PubMed] [Google Scholar]
  • 54.Aebersold R, Agar JN, Amster IJ, Baker MS, Bertozzi CR, Boja ES, Costello CE, Cravatt BF, Fenselau C, Garcia BA, et al. How many human proteoforms are there? Nat Chem Biol. 2018;14:206–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cohen P The origins of protein phosphorylation. Nat Cell Biol. 2002;4:E127–30. [DOI] [PubMed] [Google Scholar]
  • 56.Stuart T and Satija R. Integrative single-cell analysis. Nature Reviews Genetics. 2019;20:257–272. [DOI] [PubMed] [Google Scholar]
  • 57.Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, Satija R and Smibert P. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017;14:865–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fernandez DM, Rahman AH, Fernandez NF, Chudnovskiy A, Amir E-AD, Amadori L, Khan NS, Wong CK, Shamailova R, Hill CA, et al. Single-cell immune landscape of human atherosclerotic plaques. Nature medicine. 2019;25:1576–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Slavov N Unpicking the proteome in single cells. Science (New York, NY). 2020;367:512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pelisek J, Hegenloh R, Bauer S, Metschl S, Pauli J, Glukha N, Busch A, Reutersberg B, Kallmayer M, Trenner M, et al. Biobanking: Objectives, Requirements, and Future Challenges-Experiences from the Munich Vascular Biobank. J Clin Med. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Perisic L, Aldi S, Sun Y, Folkersen L, Razuvaev A, Roy J, Lengquist M, Akesson S, Wheelock CE, Maegdefessel L, et al. Gene expression signatures, pathways and networks in carotid atherosclerosis. J Intern Med. 2016;279:293–308. [DOI] [PubMed] [Google Scholar]
  • 62.Matic LP, Jesus Iglesias M, Vesterlund M, Lengquist M, Hong MG, Saieed S, Sanchez-Rivera L, Berg M, Razuvaev A, Kronqvist M, et al. Novel Multiomics Profiling of Human Carotid Atherosclerotic Plaques and Plasma Reveals Biliverdin Reductase B as a Marker of Intraplaque Hemorrhage. JACC Basic Transl Sci. 2018;3:464–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Coffey S, Lewandowski AJ, Garratt S, Meijer R, Lynum S, Bedi R, Paterson J, Yaqub M, Noble JA, Neubauer S, et al. Protocol and quality assurance for carotid imaging in 100,000 participants of UK Biobank: development and assessment. Eur J Prev Cardiol. 2017;24:1799–1806. [DOI] [PubMed] [Google Scholar]
  • 64.Malsagova K, Kopylov A, Stepanov A, Butkova T, Sinitsyna A, Izotov A and Kaysheva A. Biobanks-A Platform for Scientific and Biomedical Research. Diagnostics (Basel). 2020;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Murray CW and Rees DC. The rise of fragment-based drug discovery. Nat Chem. 2009;1:187–92. [DOI] [PubMed] [Google Scholar]
  • 66.Sanz-Nogues C and O’Brien T. In vitro models for assessing therapeutic angiogenesis. Drug Discov Today. 2016;21:1495–1503. [DOI] [PubMed] [Google Scholar]
  • 67.Staton CA, Stribbling SM, Tazzyman S, Hughes R, Brown NJ and Lewis CE. Current methods for assaying angiogenesis in vitro and in vivo. Int J Exp Pathol. 2004;85:233–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Quent VM, Loessner D, Friis T, Reichert JC and Hutmacher DW. Discrepancies between metabolic activity and DNA content as tool to assess cell proliferation in cancer research. Journal of cellular and molecular medicine. 2010;14:1003–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Stryker ZI, Rajabi M, Davis PJ and Mousa SA. Evaluation of Angiogenesis Assays. Biomedicines. 2019;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Prosser HC, Tan JT, Dunn LL, Patel S, Vanags LZ, Bao S, Ng MK and Bursill CA. Multifunctional regulation of angiogenesis by high-density lipoproteins. Cardiovasc Res. 2014;101:145–54. [DOI] [PubMed] [Google Scholar]
  • 71.Goodwin AM. In vitro assays of angiogenesis for assessment of angiogenic and anti-angiogenic agents. Microvascular research. 2007;74:172–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Rahim S and Uren A. A real-time electrical impedance based technique to measure invasion of endothelial cell monolayer by cancer cells. Journal of visualized experiments : JoVE. 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Szulcek R, Bogaard HJ and van Nieuw Amerongen GP. Electric cell-substrate impedance sensing for the quantification of endothelial proliferation, barrier function, and motility. Journal of visualized experiments : JoVE. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gagnon E, Cattaruzzi P, Griffith M, Muzakare L, LeFlao K, Faure R, Beliveau R, Hussain SN, Koutsilieris M and Doillon CJ. Human vascular endothelial cells with extended life spans: in vitro cell response, protein expression, and angiogenesis. Angiogenesis. 2002;5:21–33. [DOI] [PubMed] [Google Scholar]
  • 75.Donovan D, Brown NJ, Bishop ET and Lewis CE. Comparison of three in vitro human ‘angiogenesis’ assays with capillaries formed in vivo. Angiogenesis. 2001;4:113–21. [DOI] [PubMed] [Google Scholar]
  • 76.Hirashima M, Kataoka H, Nishikawa S, Matsuyoshi N and Nishikawa S. Maturation of embryonic stem cells into endothelial cells in an in vitro model of vasculogenesis. Blood. 1999;93:1253–63. [PubMed] [Google Scholar]
  • 77.Risau W, Sariola H, Zerwes HG, Sasse J, Ekblom P, Kemler R and Doetschman T. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development. 1988;102:471–8. [DOI] [PubMed] [Google Scholar]
  • 78.Wang S, Li X, Parra M, Verdin E, Bassel-Duby R and Olson EN. Control of endothelial cell proliferation and migration by VEGF signaling to histone deacetylase 7. Proceedings of the National Academy of Sciences. 2008;105:7738–7743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Feraud O, Cao Y and Vittet D. Embryonic stem cell-derived embryoid bodies development in collagen gels recapitulates sprouting angiogenesis. Lab Invest. 2001;81:1669–81. [DOI] [PubMed] [Google Scholar]
  • 80.Hermant B, Desroches-Castan A, Dubessay ML, Prandini MH, Huber P and Vittet D. Development of a one-step embryonic stem cell-based assay for the screening of sprouting angiogenesis. BMC Biotechnol. 2007;7:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nakagami H, Nakagawa N, Takeya Y, Kashiwagi K, Ishida C, Hayashi S, Aoki M, Matsumoto K, Nakamura T, Ogihara T, et al. Model of vasculogenesis from embryonic stem cells for vascular research and regenerative medicine. Hypertension (Dallas, Tex : 1979). 2006;48:112–9. [DOI] [PubMed] [Google Scholar]
  • 82.Feraud O, Prandini MH and Vittet D. Vasculogenesis and angiogenesis from in vitro differentiation of mouse embryonic stem cells. Methods Enzymol. 2003;365:214–28. [DOI] [PubMed] [Google Scholar]
  • 83.Kraehenbuehl TP, Ferreira LS, Hayward AM, Nahrendorf M, van der Vlies AJ, Vasile E, Weissleder R, Langer R and Hubbell JA. Human embryonic stem cell-derived microvascular grafts for cardiac tissue preservation after myocardial infarction. Biomaterials. 2011;32:1102–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang ZZ, Au P, Chen T, Shao Y, Daheron LM, Bai H, Arzigian M, Fukumura D, Jain RK and Scadden DT. Endothelial cells derived from human embryonic stem cells form durable blood vessels in vivo. Nat Biotechnol. 2007;25:317–8. [DOI] [PubMed] [Google Scholar]
  • 85.Takahashi K and Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76. [DOI] [PubMed] [Google Scholar]
  • 86.Bezenah JR, Kong YP and Putnam AJ. Evaluating the potential of endothelial cells derived from human induced pluripotent stem cells to form microvascular networks in 3D cultures. Sci Rep. 2018;8:2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Churko JM, Burridge PW and Wu JC. Generation of human iPSCs from human peripheral blood mononuclear cells using non-integrative Sendai virus in chemically defined conditions. Methods Mol Biol. 2013;1036:81–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zheng GX, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017;8:14049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Klein D iPSCs-based generation of vascular cells: reprogramming approaches and applications. Cell Mol Life Sci. 2018;75:1411–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Taura D, Sone M, Homma K, Oyamada N, Takahashi K, Tamura N, Yamanaka S and Nakao K. Induction and isolation of vascular cells from human induced pluripotent stem cells--brief report. Arteriosclerosis, thrombosis, and vascular biology. 2009;29:1100–3. [DOI] [PubMed] [Google Scholar]
  • 91.Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D, Ding BS, Schachterle W, Liu Y, Rosenwaks Z, et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell. 2013;26:204–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Clayton ZE, Tan RP, Miravet MM, Lennartsson K, Cooke JP, Bursill CA, Wise SG and Patel S. Induced pluripotent stem cell-derived endothelial cells promote angiogenesis and accelerate wound closure in a murine excisional wound healing model. Biosci Rep. 2018;38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Clayton ZE, Yuen GS, Sadeghipour S, Hywood JD, Wong JW, Huang NF, Ng MK, Cooke JP and Patel S. A comparison of the pro-angiogenic potential of human induced pluripotent stem cell derived endothelial cells and induced endothelial cells in a murine model of peripheral arterial disease. Int J Cardiol. 2017;234:81–89. [DOI] [PubMed] [Google Scholar]
  • 94.Belair DG, Whisler JA, Valdez J, Velazquez J, Molenda JA, Vickerman V, Lewis R, Daigh C, Hansen TD, Mann DA, et al. Human vascular tissue models formed from human induced pluripotent stem cell derived endothelial cells. Stem Cell Rev Rep. 2015;11:511–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Palladino A, Mavaro I, Pizzoleo C, De Felice E, Lucini C, de Girolamo P, Netti PA and Attanasio C. Induced Pluripotent Stem Cells as Vasculature Forming Entities. J Clin Med. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kurokawa YK, Yin RT, Shang MR, Shirure VS, Moya ML and George SC. Human Induced Pluripotent Stem Cell-Derived Endothelial Cells for Three-Dimensional Microphysiological Systems. Tissue Eng Part C Methods. 2017;23:474–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zanotelli MR, Ardalani H, Zhang J, Hou Z, Nguyen EH, Swanson S, Nguyen BK, Bolin J, Elwell A, Bischel LL, et al. Stable engineered vascular networks from human induced pluripotent stem cell-derived endothelial cells cultured in synthetic hydrogels. Acta Biomater. 2016;35:32–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Rodríguez-Rodríguez DR, Ramírez-Solís R, Garza-Elizondo MA, Garza-Rodríguez ML and Barrera-Saldaña HA. Genome editing: A perspective on the application of CRISPR/Cas9 to study human diseases (Review). Int J Mol Med. 2019;43:1559–1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Warren CR, O’Sullivan JF, Friesen M, Becker CE, Zhang X, Liu P, Wakabayashi Y, Morningstar JE, Shi X, Choi J, et al. Induced Pluripotent Stem Cell Differentiation Enables Functional Validation of GWAS Variants in Metabolic Disease. Cell Stem Cell. 2017;20:547–557.e7. [DOI] [PubMed] [Google Scholar]
  • 100.Irvin MW, Zijlstra A, Wikswo JP and Pozzi A. Techniques and assays for the study of angiogenesis. Exp Biol Med (Maywood). 2014;239:1476–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Burbridge MF and West DC. Rat Aortic Ring : 3D Model of Angiogenesis In Vitro. Methods Mol Med. 2001;46:185–204. [DOI] [PubMed] [Google Scholar]
  • 102.Nicosia RF and Ottinetti A. Growth of microvessels in serum-free matrix culture of rat aorta. A quantitative assay of angiogenesis in vitro. Lab Invest. 1990;63:115–22. [PubMed] [Google Scholar]
  • 103.Grebenyuk S and Ranga A. Engineering Organoid Vascularization. Front Bioeng Biotechnol. 2019;7:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Clevers H Modeling Development and Disease with Organoids. Cell. 2016;165:1586–1597. [DOI] [PubMed] [Google Scholar]
  • 105.Duttenhoefer F, Lara de Freitas R, Meury T, Loibl M, Benneker LM, Richards RG, Alini M and Verrier S. 3D scaffolds co-seeded with human endothelial progenitor and mesenchymal stem cells: evidence of prevascularisation within 7 days. Eur Cell Mater. 2013;26:49–64; discussion 64–5. [DOI] [PubMed] [Google Scholar]
  • 106.Shah S and Kang KT. Two-Cell Spheroid Angiogenesis Assay System Using Both Endothelial Colony Forming Cells and Mesenchymal Stem Cells. Biomol Ther (Seoul). 2018;26:474–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zucchelli E, Majid QA and Foldes G. New artery of knowledge: 3D models of angiogenesis. Vasc Biol. 2019;1:H135–H143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Korff T and Augustin HG. Integration of endothelial cells in multicellular spheroids prevents apoptosis and induces differentiation. J Cell Biol. 1998;143:1341–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wörsdörfer P, Dalda N, Kern A, Krüger S, Wagner N, Kwok CK, Henke E and Ergün S. Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells. Scientific Reports. 2019;9:15663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.van Duinen V, Stam W, Mulder E, Famili F, Reijerkerk A, Vulto P, Hankemeier T and van Zonneveld AJ. Robust and Scalable Angiogenesis Assay of Perfused 3D Human iPSC-Derived Endothelium for Anti-Angiogenic Drug Screening. International journal of molecular sciences. 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Dikici S, Claeyssens F and MacNeil S. Bioengineering Vascular Networks to Study Angiogenesis and Vascularization of Physiologically Relevant Tissue Models in Vitro. ACS Biomater Sci Eng. 2020;6:3513–3528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ji S, Almeida E and Guvendiren M. 3D bioprinting of complex channels within cell-laden hydrogels. Acta Biomater. 2019;95:214–224. [DOI] [PubMed] [Google Scholar]
  • 113.Mastrullo V, Cathery W, Velliou E, Madeddu P and Campagnolo P. Angiogenesis in Tissue Engineering: As Nature Intended? Front Bioeng Biotechnol. 2020;8:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kargozar S, Baino F, Hamzehlou S, Hamblin MR and Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev. 2020;49:5008–5057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Abdalla AME, Xiao L, Ullah MW, Yu M, Ouyang C and Yang G. Current Challenges of Cancer Anti-angiogenic Therapy and the Promise of Nanotherapeutics. Theranostics. 2018;8:533–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vargas GE, Haro Durand LA, Cadena V, Romero M, Mesones RV, Mackovic M, Spallek S, Spiecker E, Boccaccini AR and Gorustovich AA. Effect of nano-sized bioactive glass particles on the angiogenic properties of collagen based composites. J Mater Sci Mater Med. 2013;24:1261–9. [DOI] [PubMed] [Google Scholar]
  • 117.Darweesh RS, Ayoub NM and Nazzal S. Gold nanoparticles and angiogenesis: molecular mechanisms and biomedical applications. Int J Nanomedicine. 2019;14:7643–7663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kommareddy S and Amiji M. Antiangiogenic gene therapy with systemically administered sFlt-1 plasmid DNA in engineered gelatin-based nanovectors. Cancer Gene Ther. 2007;14:488–98. [DOI] [PubMed] [Google Scholar]
  • 119.Montero RB, Vial X, Nguyen DT, Farhand S, Reardon M, Pham SM, Tsechpenakis G and Andreopoulos FM. bFGF-containing electrospun gelatin scaffolds with controlled nano-architectural features for directed angiogenesis. Acta Biomater. 2012;8:1778–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wagner S, Rothweiler F, Anhorn MG, Sauer D, Riemann I, Weiss EC, Katsen-Globa A, Michaelis M, Cinatl J Jr., Schwartz D, Kreuter J, et al. Enhanced drug targeting by attachment of an anti alphav integrin antibody to doxorubicin loaded human serum albumin nanoparticles. Biomaterials. 2010;31:2388–98. [DOI] [PubMed] [Google Scholar]
  • 121.Rajagopalan S, Trachtenberg J, Mohler E, Olin J, McBride S, Pak R, Rasmussen H and Crystal R. Phase I study of direct administration of a replication deficient adenovirus vector containing the vascular endothelial growth factor cDNA (CI-1023) to patients with claudication. Am J Cardiol. 2002;90:512–6. [DOI] [PubMed] [Google Scholar]
  • 122.Iyer SR and Annex BH. Therapeutic Angiogenesis for Peripheral Artery Disease: Lessons Learned in Translational Science. JACC Basic Transl Sci. 2017;2:503–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Ding X, Su Y, Wang C, Zhang F, Chen K, Wang Y, Li M and Wang W. Synergistic Suppression of Tumor Angiogenesis by the Co-delivering of Vascular Endothelial Growth Factor Targeted siRNA and Candesartan Mediated by Functionalized Carbon Nanovectors. ACS Appl Mater Interfaces. 2017;9:23353–23369. [DOI] [PubMed] [Google Scholar]
  • 124.Masotti A, Miller MR, Celluzzi A, Rose L, Micciulla F, Hadoke PW, Bellucci S and Caporali A. Regulation of angiogenesis through the efficient delivery of microRNAs into endothelial cells using polyamine-coated carbon nanotubes. Nanomedicine. 2016;12:1511–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Pacelli S, Acosta F, Chakravarti AR, Samanta SG, Whitlow J, Modaresi S, Ahmed RPH, Rajasingh J and Paul A. Nanodiamond-based injectable hydrogel for sustained growth factor release: Preparation, characterization and in vitro analysis. Acta Biomater. 2017;58:479–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang Z, Niu B, Chen J, He X, Bao X, Zhu J, Yu H and Li Y. The use of lipid-coated nanodiamond to improve bioavailability and efficacy of sorafenib in resisting metastasis of gastric cancer. Biomaterials. 2014;35:4565–72. [DOI] [PubMed] [Google Scholar]
  • 127.McLuckie M, Schmidt CA, Oosthuysen A, Sanchez-Macedo N, Merker H, Bezuidenhout D, Hoerstrup SP and Lindenblatt N. High heparin content surface-modified polyurethane discs promote rapid and stable angiogenesis in full thickness skin defects through VEGF immobilization. J Biomed Mater Res A. 2017;105:2543–2550. [DOI] [PubMed] [Google Scholar]
  • 128.Sahoo N, Sahoo RK, Biswas N, Guha A and Kuotsu K. Recent advancement of gelatin nanoparticles in drug and vaccine delivery. Int J Biol Macromol. 2015;81:317–31. [DOI] [PubMed] [Google Scholar]
  • 129.Flores AM, Ye J, Jarr KU, Hosseini-Nassab N, Smith BR and Leeper NJ. Nanoparticle Therapy for Vascular Diseases. Arterioscler Thromb Vasc Biol. 2019;39:635–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lau P, Bidin N, Islam S, Shukri W, Zakaria N, Musa N and Krishnan G. Influence of gold nanoparticles on wound healing treatment in rat model: Photobiomodulation therapy. Lasers Surg Med. 2017;49:380–386. [DOI] [PubMed] [Google Scholar]
  • 131.Liu W, Zhang G, Wu J, Zhang Y, Liu J, Luo H and Shao L. Insights into the angiogenic effects of nanomaterials: mechanisms involved and potential applications. J Nanobiotechnology. 2020;18:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Faria M, Bjornmalm M, Thurecht KJ, Kent SJ, Parton RG, Kavallaris M, Johnston APR, Gooding JJ, Corrie SR, Boyd BJ, et al. Minimum information reporting in bio-nano experimental literature. Nat Nanotechnol. 2018;13:777–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Boada C, Zinger A, Tsao C, Zhao P, Martinez Jonathan O, Hartman K, Naoi T, Sukhovershin R, Sushnitha M, Molinaro R, et al. Rapamycin-Loaded Biomimetic Nanoparticles Reverse Vascular Inflammation. Circ Res. 2020;126:25–37. [DOI] [PubMed] [Google Scholar]
  • 134.Leeper NJ, Lee JT and Cooke JP. Two decades of progress in vascular medicine. Am J Med. 2011;124:791–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Niiyama H, Huang NF, Rollins MD and Cooke JP. Murine model of hindlimb ischemia. J Vis Exp. 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lotfi S, Patel AS, Mattock K, Egginton S, Smith A and Modarai B. Towards a more relevant hind limb model of muscle ischaemia. Atherosclerosis. 2013;227:1–8. [DOI] [PubMed] [Google Scholar]
  • 137.Long CA, Timmins LH, Koutakis P, Goodchild TT, Lefer DJ, Pipinos II, Casale GP and Brewster LP. An endovascular model of ischemic myopathy from peripheral arterial disease. J Vasc Surg. 2017;66:891–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Krishna SM, Omer SM, Li J, Morton SK, Jose RJ and Golledge J. Development of a two-stage limb ischemia model to better simulate human peripheral artery disease. Sci Rep. 2020;10:3449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Helisch A, Wagner S, Khan N, Drinane M, Wolfram S, Heil M, Ziegelhoeffer T, Brandt U, Pearlman JD, Swartz HM, et al. Impact of mouse strain differences in innate hindlimb collateral vasculature. Arterioscler Thromb Vasc Biol. 2006;26:520–6. [DOI] [PubMed] [Google Scholar]
  • 140.Robinet P, Milewicz DM, Cassis LA, Leeper NJ, Lu HS and Smith JD. Consideration of Sex Differences in Design and Reporting of Experimental Arterial Pathology Studies-Statement From ATVB Council. Arterioscler Thromb Vasc Biol. 2018;38:292–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kullo IJ and Leeper NJ. The genetic basis of peripheral arterial disease: current knowledge, challenges, and future directions. Circ Res. 2015;116:1551–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Winkel LC, Hoogendoorn A, Xing R, Wentzel JJ and Van der Heiden K. Animal models of surgically manipulated flow velocities to study shear stress-induced atherosclerosis. Atherosclerosis. 2015;241:100–10. [DOI] [PubMed] [Google Scholar]
  • 143.Chen YC, Bui AV, Diesch J, Manasseh R, Hausding C, Rivera J, Haviv I, Agrotis A, Htun NM, Jowett J, et al. A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling. Circ Res. 2013;113:252–65. [DOI] [PubMed] [Google Scholar]
  • 144.Cheng C, Tempel D, van Haperen R, van der Baan A, Grosveld F, Daemen MJ, Krams R and de Crom R. Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress. Circulation. 2006;113:2744–53. [DOI] [PubMed] [Google Scholar]
  • 145.Sasaki T, Kuzuya M, Nakamura K, Cheng XW, Shibata T, Sato K and Iguchi A. A simple method of plaque rupture induction in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2006;26:1304–9. [DOI] [PubMed] [Google Scholar]
  • 146.von der Thusen JH, van Berkel TJ and Biessen EA. Induction of rapid atherogenesis by perivascular carotid collar placement in apolipoprotein E-deficient and low-density lipoprotein receptor-deficient mice. Circulation. 2001;103:1164–70. [DOI] [PubMed] [Google Scholar]
  • 147.Hartwig H, Silvestre-Roig C, Hendrikse J, Beckers L, Paulin N, Van der Heiden K, Braster Q, Drechsler M, Daemen MJ, Lutgens E, et al. Atherosclerotic Plaque Destabilization in Mice: A Comparative Study. PLoS One. 2015;10:e0141019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Silvestre-Roig C, de Winther MP, Weber C, Daemen MJ, Lutgens E and Soehnlein O. Atherosclerotic plaque destabilization: mechanisms, models, and therapeutic strategies. Circ Res. 2014;114:214–26. [DOI] [PubMed] [Google Scholar]
  • 149.Wang Y, Nanda V, Direnzo D, Ye J, Xiao S, Kojima Y, Howe KL, Jarr KU, Flores AM, Tsantilas P, et al. Clonally expanding smooth muscle cells promote atherosclerosis by escaping efferocytosis and activating the complement cascade. Proc Natl Acad Sci U S A. 2020;117:15818–15826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wang Y, Dubland JA, Allahverdian S, Asonye E, Sahin B, Jaw JE, Sin DD, Seidman MA, Leeper NJ and Francis GA. Smooth Muscle Cells Contribute the Majority of Foam Cells in ApoE (Apolipoprotein E)-Deficient Mouse Atherosclerosis. Arterioscler Thromb Vasc Biol. 2019;39:876–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.DiRenzo D, Owens GK and Leeper NJ. “Attack of the Clones”: Commonalities Between Cancer and Atherosclerosis. Circ Res. 2017;120:624–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Jarr KU, Ye J, Kojima Y, Nanda V, Flores AM, Tsantilas P, Wang Y, Hosseini-Nassab N, Eberhard AV, Lotfi M, et al. (18)F-Fluorodeoxyglucose-Positron Emission Tomography Imaging Detects Response to Therapeutic Intervention and Plaque Vulnerability in a Murine Model of Advanced Atherosclerotic Disease-Brief Report. Arterioscler Thromb Vasc Biol. 2020;40:2821–2828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Huynh P, Phie J, Krishna SM and Golledge J. Systematic review and meta-analysis of mouse models of diabetes-associated ulcers. BMJ Open Diabetes Res Care. 2020;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Murabito JM, Evans JC, Nieto K, Larson MG, Levy D and Wilson PW. Prevalence and clinical correlates of peripheral arterial disease in the Framingham Offspring Study. Am Heart J. 2002;143:961–5. [DOI] [PubMed] [Google Scholar]
  • 155.Dunn L, Prosser HC, Tan JT, Vanags LZ, Ng MK and Bursill CA. Murine model of wound healing. Journal of visualized experiments : JoVE. 2013:e50265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chen JS, Longaker MT and Gurtner GC. Murine models of human wound healing. Methods Mol Biol. 2013;1037:265–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Galiano RD, Michaels Jt, Dobryansky M, Levine JP and Gurtner GC. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004;12:485–92. [DOI] [PubMed] [Google Scholar]
  • 158.Wassermann E, van Griensven M, Gstaltner K, Oehlinger W, Schrei K and Redl H. A chronic pressure ulcer model in the nude mouse. Wound Repair Regen. 2009;17:480–4. [DOI] [PubMed] [Google Scholar]
  • 159.Masson-Meyers DS, Andrade TAM, Caetano GF, Guimaraes FR, Leite MN, Leite SN and Frade MAC. Experimental models and methods for cutaneous wound healing assessment. Int J Exp Pathol. 2020;101:21–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Thanigaimani S, Phie J and Golledge J. Animal models of ischemic limb ulcers: a systematic review and meta-analysis. BMJ Open Diabetes Res Care. 2020;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Alizadeh N, Pepper MS, Modarressi A, Alfo K, Schlaudraff K, Montandon D, Gabbiani G, Bochaton-Piallat ML and Pittet B. Persistent ischemia impairs myofibroblast development in wound granulation tissue: a new model of delayed wound healing. Wound Repair Regen. 2007;15:809–16. [DOI] [PubMed] [Google Scholar]
  • 162.Andre-Levigne D, Modarressi A, Pignel R, Bochaton-Piallat ML and Pittet-Cuenod B. Hyperbaric oxygen therapy promotes wound repair in ischemic and hyperglycemic conditions, increasing tissue perfusion and collagen deposition. Wound Repair Regen. 2016;24:954–965. [DOI] [PubMed] [Google Scholar]
  • 163.Tobalem M, Levigne D, Modarressi A, Atashi F, Villard F, Hinz B and Pittet-Cuenod B. Hyperglycemia Interacts with Ischemia in a Synergistic Way on Wound Repair and Myofibroblast Differentiation. Plast Reconstr Surg Glob Open. 2015;3:e471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Ross EG, Shah NH, Dalman RL, Nead KT, Cooke JP and Leeper NJ. The use of machine learning for the identification of peripheral artery disease and future mortality risk. J Vasc Surg. 2016;64:1515–1522.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Villarreal MF, Siracuse JJ, Menard M, Assmann SF, Siami FS, Rosenfield K, Strong MB and Farber A. Enrollment Obstacles in a Randomized Controlled Trial: A Performance Survey of Enrollment in BEST-CLI Sites. Ann Vasc Surg. 2020;62:406–411. [DOI] [PubMed] [Google Scholar]
  • 166.Allan RB, Vun SV and Spark JI. A Comparison of Measures of Endothelial Function in Patients with Peripheral Arterial Disease and Age and Gender Matched Controls. Int J Vasc Med. 2016;2016:2969740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chowdhury MM, Tarkin JM, Albaghdadi MS, Evans NR, Le EPV, Berrett TB, Sadat U, Joshi FR, Warburton EA, Buscombe JR, et al. Vascular Positron Emission Tomography and Restenosis in Symptomatic Peripheral Arterial Disease: A Prospective Clinical Study. JACC Cardiovasc Imaging. 2020;13:1008–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES