Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Circulation Research

PERIPHERAL VASCULAR DISEASE COMPENDIUM

New Directions in Therapeutic Angiogenesis and


Arteriogenesis in Peripheral Arterial Disease
Brian H. Annex , John P. Cooke

ABSTRACT: The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a
significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the
large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired
functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation.
Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite
advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms,
and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive
disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease.
Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited
benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein
convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic
ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have
included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell
therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups
and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned
to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic
Downloaded from http://ahajournals.org by on July 26, 2023

angiogenesis and arteriogenesis for PAD.

Key Words: cytokines ◼ metabolism ◼ microvessels ◼ permeability ◼ stem cells

THE PERVASIVE AND PERSISTENT overlap those of coronary artery and cerebrovascular
disease, diabetes and smoking have been and remain
PROBLEMS OF PERIPHERAL ARTERIAL the 2 strongest age-adjusted risk factors for the devel-
DISEASE opment of PAD.1,5,7 Patients with PAD continue to suf-
Systemic atherosclerosis remains the number one fer excessively high risks of heart attack and stroke
cause of morbidity and mortality in the western world when compared with patients without PAD.8–10
and is the major cause of peripheral arterial dis- The 2 major symptomatic manifestations of PAD are
ease (PAD), causing obstructions in blood flow in intermittent claudication (IC) and critical limb ischemia
one or more of the major leg arteries.1–4 Estimates (CLI).1,8,11 IC is the most prevalent clinical manifestation in
place the prevalence of PAD at over 14 million in the patients with PAD and is defined by the presence of calf
United States and >200 million people worldwide.1–3,5 or buttock muscle discomfort produced by exercise that is
In patients, PAD is still defined by the finding of an relieved by rest.12 CLI is the most severe manifestation of
ankle-brachial blood pressure index of 0.90 or less.5–7 PAD and is characterized by one or more of the following:
Although the risk factors for the development of PAD leg pain occurring at rest, ulceration, and gangrene.11,13

Correspondence to: Brian H. Annex, MD, J. Harold Harrison, MD, Joseph C. “Rusty” Walter and Carol Walter Looke Presidential Distinguised Chair, Chair,
Department of Medicine, Medical College of Georgia, Augusta University, 1120 15th St BI 5076, Augusta, GA 30912, Email bannex@augusta.edu or John P.
Cooke, MD, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave, Mail Stop R10-South, Houston, TX 77030, Email
jpcooke@houstonmethodist.org
For Sources of Funding and Disclosures, see page 1953.
© 2021 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

1944   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

with PAD are similar to the impairment seen in patients

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
Nonstandard Abbreviations and Acronyms with advanced congestive heart failure.14,15
Currently, medical therapies used to treat patients
ACE angiotensin-converting enzyme with PAD include antiplatelet and antithrombotic agents,
ARB angiotensin receptor blocker statins to lower cholesterol, antihypertensive therapy
CCR2 C-C chemokine receptor 2 with ACE (angiotensin-converting enzyme) inhibitors or
CLI critical limb ischemia ARB (angiotensin receptor blocker) or β-blockers, blood
CXCR-4 C-X-C chemokine receptor type 4 glucose control, and smoking cessation. These thera-
Del-1 developmental endothelial locus 1
pies were largely established from treatments used for
patients with coronary artery disease. In patients with
EC endothelial cell
PAD, these agents may prevent heart attack and stroke,
eNOS endothelial nitric oxide synthase
but clinical cardiovascular event rates in patients with
FGF fibroblast growth factor PAD remain high, and even new anticoagulant therapies
G-CSF granulocyte-colony stimulating factor and intensive lipid-lowering therapies have not consis-
GM-CSF granulocyte macrophage-colony stimu- tently altered the progression of disabling claudication
lating factor or limb loss.1,9,10 These medical therapies do not improve
HGF hepatocyte growth factor lower limb perfusion in patients with PAD.16 Surgical or
HIF-1 hypoxia-inducible factor 1 endovascular procedures may be suboptimal in reliev-
HLI hindlimb ischemia ing symptoms and may not be indicated because of the
HSS hypoxia and serum starvation extent of disease or because of comorbid conditions.
IC intermittent claudication Finally, even after successful large vessel revasculariza-
ICAM intercellular adhesion molecule tion, residual microvascular disease may well limit the
IL interleukin effectiveness of therapies in patients with PAD.11,13,16 Still
iNOS inducible nitric oxide synthase
today PAD effects both legs and life. Few areas in cardio-
vascular medicine are more in need of new therapeutic
iPSCs induced pluripotent stem cells
modalities than PAD.
lncRNA long noncoding RNA
PAD peripheral arterial disease
PCSK-9 proprotein convertase subtilisin/kexin
ANGIOGENESIS, ARTERIOGENESIS,
Downloaded from http://ahajournals.org by on July 26, 2023

type 9
PDGF platelet-derived growth factor VASCULOGENESIS, AND PAD
PFK-1 phosphofructokinase 1 Within the adult, the overriding purpose of the vasculature
SDF-1 stromal-derived factor 1 is to regulate blood flow (delivery of oxygen and nutrients
TNF tumor necrosis factor along with the removal of toxins) to organs to meet the
VEGF vascular endothelial growth factor metabolic demands of active cells. The central problem in
PAD is the limitation of perfusion to the muscle and soft
tissue of the leg(s). It would, therefore, seem rational to
Of critical importance, classic claudication symptoms attempt to improve perfusion in PAD by enhancing vascu-
actually occur in only a minority of patients with PAD, lar regeneration and function. Angiogenesis is defined as
and this has been shown in those with a confirmed low the growth and proliferation of blood vessels from exist-
ankle-brachial blood pressure index.6,7,10 Despite its high ing vascular structures.17 Angiogenesis is, of course, criti-
disease prevalence, the low ankle-brachial blood pres- cal in embryonic development. In adults, angiogenesis is
sure index is not sufficiently used, and because symp- required for tissue repair or remodeling, as well as resto-
toms may be atypical or absent in many patients, PAD ration of tissue perfusion.18 Angiogenesis can be physi-
often goes unrecognized and undertreated.2–4 Of critical ologically regulated such as in the response to exercise
importance is that all patients with PAD, even those who training. A major process in angiogenesis is the sprout-
lack symptoms, have significant disability with limita- ing of endothelial cells (ECs) from preexisting capillaries
tions in walking capacity, daily functional activities, and under the regulation of angiogenic factors such as VEGF
exercise performance.14,15 The clinical course of patients (vascular endothelial growth factor) generated by isch-
with PAD is strikingly different based upon whether the emic tissue (Figure 1). The sprouting ECs migrate, pro-
patients present with IC or CLI. Patients with CLI suffer liferate, and form lumens while other processes that may
rates of death and major amputation of 30% or more by help to generate a functional microvasculature include
1-year, whereas over a 5-year period, only 1.0% to 3.3% intussusception of existing capillaries,17,18 the incorpo-
of IC patients will require amputation.1,8–10 Even in the ration of circulating endothelial progenitors,19,20 and the
absence of CLI, the functional limitations seen in patients generation of cytokines by circulating angiogenic cells.21

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1945


Annex and Cooke Vascular Regeneration in PAD

Pathological angiogenesis may support another


DISEASE COMPENDIUM
PERIPHERAL VASCULAR

pathological process and examples would include


tumor angiogenesis, atherosclerotic plaque neovas-
cularization, or age-related macular degeneration. In
each of these tobacco-related conditions, nicotinic
acetylcholine receptors on the endothelium are stimu-
lated by exogenous nicotine to proliferate, migrate, and
form a microvasculature that supports the underlying
pathological process.22,23 Another form of pathologi-
cal angiogenesis is when the angiogenic response to
ischemia is inadequate to meet tissue demands, and
this most commonly occurs in the settings of coronary
artery disease and PAD. Here, hypoxia in heart and
leg muscles due to poor tissue perfusion activates the
transcription factor HIF-1 (hypoxia-inducible factor 1)
as its subunit HIF-1α has a degradation domain that
is dependent upon oxygen tension.24 With low oxygen
levels, HIF-1α is stabilized, translocates to the nucleus,
and activates target genes including VEGF, FGF (fibro-
blast growth factor), HGF (hepatocyte growth factor),
PDGF (platelet-derived growth factor), Del-1 (develop-
mental endothelial locus 1), angiopoietins, and matrix
metalloproteinases to facilitate angiogenesis.25 Notably,
exogenous administration of each of these genes or the
proteins that they encode, have been shown to improve
limb blood flow in animal models of PAD,26–31 but as
discussed below, have failed in randomized clinical trials
to significantly ameliorate PAD.
The remodeling of preexisting collateral channels is
Downloaded from http://ahajournals.org by on July 26, 2023

termed arteriogenesis. In their normal state, these col-


lateral channels are narrow, high resistance vessels, and
they provide little blood flow to their distal tissue bed.
However, when a major conduit becomes obstructed,
blood flow is redirected through the collateral channels
(Figure 1) which causes alterations in vascular wall shear
stress. This hemodynamic stimulus provokes an increase
in the diameter and wall thickness of the collateral chan-
nels, with proliferation of vascular cells and turnover of
the vascular matrix.32,33 Other factors involved in arterio-
genesis include chemokines and adhesion molecules,
such as ICAM (intercellular adhesion molecule), CCR2
(C-C chemokine receptor 2), Delta-like 1, SDF-1 (stro-
mal-derived factor 1), which recruit monocytes that then
Figure 1. Processes to restore perfusion in peripheral arterial facilitate the positive remodeling by elaborating metal-
disease (PAD). loproteinases and cytokines..34–37 Whereas macrophages
In angiogenesis, new microvasculature is derived from the sprouting are the cell most linked to arteriogenesis, recent data
of preexisting capillaries under the influence of VEGF (vascular have also linked inflammatory (referred to as M1-like)
endothelial growth factor), FGF (fibroblast growth factor), and other
angiogenic cytokines. Arteriogenesis is the positive remodeling
of preexisting collaterals when blood flow is redirected through Figure 1 Continued. that is expressed by the ischemic tissue.
these channels. The positive remodeling of these channels is due, Most CACs are of hematopoietic origin and contribute to restoration
in part, to the mobilization of monocytes by G-CSF (granulocyte- of perfusion by secreting angiogenic cytokines. Rare CACs are
colony stimulating factor) and vascular expression of monocyte of endothelial lineage and can incorporate into the newly forming
chemotactic protein, the ligand for the monocyte CCR2 (C-C vessels. Transdifferentiation of fibroblasts to endothelial cells may
chemokine receptor type 2). Circulating angiogenic cells (CACs) also contribute to angiogenesis. In the setting of inflammatory
are also mobilized by G-CSF to participate in adult vasculogenesis. signaling, increased DNA accessibility permits fluidity of cell fate,
These cells express CXCR-4 (C-X-C chemokine receptor type 4), and environmental influences such as hypoxia activate transcription
the receptor for SDF-1 (stromal-derived factor 1) (Continued ) factors determining endothelial lineage.

1946   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

macrophages in processes that negatively impact angio- NEW DIRECTIONS FOR POTENTIAL

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
genesis and which may be a target for therapeutics.38–40
Inflammatory macrophages in ischemic tissue may be a
THERAPEUTIC SOLUTIONS IN PAD
source for circulating cytokines (ie, TNF [tumor necrosis Angiogenesis Independent of VEGF
factor]-α and IL [interleukin]-6) that can trigger acute To date a substantial majority of the completed human
cardiovascular events.39,41 therapeutic angiogenesis trials, in whole or in part, sought
Adult vasculogenesis refers to the mobilization of to activate the well described, canonical, VEGF-VEGFR2/
circulating cells with angiogenic potential from the Akt-1/eNOS (endothelial nitric oxide synthase) pathway;
bone marrow into the circulation by factors, such as where increases in bioavailable nitric oxide is a critical
G-CSF (granulocyte-colony stimulating factor) and mediator.58–60 Promoting angiogenesis independent of
GM-CSF (granulocyte macrophage-colony stimulating this pathway may be important for humans with PAD as
factor). These CD34+ circulating angiogenic cells may these patients frequently have comorbid conditions (even
also express CD133, VEGFR2, and CXCR-4 (C-X-C diabetes which is a frequently causes and modifies PAD)
chemokine receptor type 4; Figure 1).42–45 Under the which can limit the generation, or bioavailability, of NO.16
influence of SDF-1 (which binds to the chemokine Preclinical models of PAD remain an invaluable tool for
receptor CXCR-4), angiogenic cells are recruited to testing the potential efficacy and bioactivity of putative
the ischemic tissue.46,47 Bone marrow-derived angio- therapeutic agents as well as to identify novel therapeu-
genic cells are largely of hematopoietic lineage and tics. Using established differences in the extent of perfu-
largely improve perfusion by secreting cytokines and sion recovery and necrosis that occurs across informative
metalloproteinases, although a rare fraction may differ- inbred mouse strains and congenic lines, Dokun et al61
entiate into mature endothelium in vivo.19,48,49 It is also were the first to identify a quantitative trait locus on the
possible that angiogenic cytokines cause mature ECs short arm of mouse chromosome 7 (termed LSq-1) that
in other nonischemic organs to be mobilized into the contained potential gene(s) that were sufficient to deter-
circulation, home to ischemic tissue, and contribute to mine the extent of perfusion recovery and tissue loss after
angiogenesis.19,50 hindlimb ischemia (HLI). This was an unbiased approach
In patients with PAD, there is some degree of and had the advantage of allowing us to look for genes
endogenous arteriogenesis (and angiogenesis), but that modified the course of PAD in the setting of compara-
the capacity of the processes even when active are ble occlusions.16,61 More importantly, this strategy had the
limited in their ability to restore limb perfusion to nor-
Downloaded from http://ahajournals.org by on July 26, 2023

advantage of not requiring that we preselect for known


mal levels.51 In patients with claudication, capillary angiogenesic or arteriogenic genes or pathways. One of
density is often reduced and is related to a patients’ the first candidates to emerge from this approach was the
functional capacity.52 Notably, an increase in capillary IL-21R (IL-21 receptor) which, resides at/very near the
density is observed in the limb skeletal muscle of PAD peak of genetic linkage to variations in perfusion recovery
patients with exercise training, which is still today the and necrosis in the mouse model of PAD and the potential
cornerstone of therapy in patients with PAD.53 Moving role of this receptor in modulating angiogenesis in the set-
from concept to the clinic, we hope to harness blood ting of PAD has supporting human data.62,63
vessel growth to improve perfusion to ischemic tis- The IL-21R was identified about 2 decades ago in
sue, an approach which has been termed therapeutic the laboratory of Dr Warren J. Leonard at the National
angiogenesis or medical revascularization. Perhaps, Institutes of Health/National Heart, Lung, and Blood
a better term is vascular regeneration, which encom- Institute and has emerged as a key molecule in innate
passes angiogenesis, arteriogenesis, and vasculo- and adaptive immunity, with IL-21 serving as its sole
genesis, the combination of which processes may be ligand.64,65 Early work with IL-21R indicated that it was
necessary for substantial benefit. Investigation into angiostatic, but these observations were obtained using
therapeutic angiogenesis dates back to the work of mouse and human ECs cultured in serum supplemented
the late Isner et al,54 who administered by intraarte- with bFGF (basic FGF, a potent angiogenic agent) under
rial infusion a plasmid vector encoding VEGF to treat normoxic conditions.66 By contrast, skeletal muscle tis-
a 70-year-old female with midfoot 3-vessel runoff sue has low levels of VEGF, bFGF, and other cytokine
occlusion and digit gangrene. Their work lacked a suf- growth factors.67,68 Accordingly, we altered the in vitro
ficient control group to draw conclusions but stimu- conditions to be more similar to those seen in isch-
lated a great number of clinical trials of therapeutic emic skeletal muscle (with ECs subjected to hypoxia
angiogenesis involving thousands of patients, with and serum starvation, ie, HSS). Under HSS conditions,
varying experimental protocols, agents, and modes human ECs upregulate IL-21R and exogenous ligand-
of delivery. Regrettably, these studies have failed to induced angiogenesis.69 The differences between the
deliver significantly positive results, as previously angiogenic outcomes from the in vitro experiments
reviewed.16,55–57 Thus, new approaches are needed for conducted under HSS versus the angiostatic outcomes
vascular regeneration in PAD. seen with ECs in growth factor rich, normoxic conditions

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1947


Annex and Cooke Vascular Regeneration in PAD

were associated with differences in downstream signal- of VEGF-A led to the formation of leaky dysfunc-
DISEASE COMPENDIUM
PERIPHERAL VASCULAR

ing. Specifically, under HSS, ligand-mediated IL-21R tional vessels, whereas the coordinated expression of
activation activated STAT3, not STAT1 nor Akt.62,69 all of the splice variants led to the formation of more
In wild-type C57BL/6 mice, we found that the IL-21R functional blood vessels in a mouse ear angiogenesis
was significantly upregulated within ischemic muscle fol- model. Dai et al75 demonstrated that a VEGF-A acti-
lowing HLI, and the increases were largely within the EC vating transcription factor could increase expression
population. By contrast, the IL-21R showed no increase of multiple VEGF isoforms in ischemic muscle in vivo
in expression in Balb/C mice, a mouse strain that devel- and promote angiogenesis following HLI. An additional
ops a sustained perfusion deficit and greater tissue loss and less appreciated splice variant occurs with alter-
following induction of HLI.62 These results suggested nate splicing of the eighth exon of VEGF-A results in
that IL-21R on ECs might play an important role in the a 6 amino acid switch that changes the pro-angiogenic
response to ischemia. The IL-21R seemed to have little VEGFxxxa (xxx =the number of amino acids) to the anti-
role in non-ECs in muscle, as it was not detectable or angiogenic VEGFxxxb (VEGF165b for 165 amino acids)
present at very low levels in vascular smooth muscle and isoform.40,76–78 This splicing can occur for any of the
skeletal muscle cells in both normoxic and HSS condi- amino acid length transcripts but has been best stud-
tions, and IL-21 administration had no effect on vascular ied for the 165 amino acid isoform.78
smooth muscle nor skeletal muscle cells survival under Kikuchi et al77 showed that peripheral blood mono-
HSS. Furthermore, IL-21 (ligand) levels do not change cytes (isolated by gradient centrifugation) from PAD
following HLI in C57Bl/6 nor Balb/C mice; in human versus control patients expressed significantly higher
umbilical vein ECs IL-21 protein was not detectable in VEGF165b by quantitative polymerase chain reaction and
cells before or after HSS.62 Silencing the IL-21R recep- then in the mouse HLI model showed that intramuscular
tor blocked the angiogenic effect of exogenous ligand, delivery of a VEGF165b specific antibody improved per-
in vitro and in vivo, and when angiogenesis occurred fusion recovery in experimental PAD.77 Their conclusion
there was no change in the Akt/eNOS pathway.62 Con- and indeed the conclusion in similar studies of the VEGF
sistent with its modulation of perfusion changes follow- system focused on the activation the VEGFR2 pathway
ing vascular injury, Lee et al70 later demonstrated that as the mechanism of action.77,79,80 Indeed, even studies
IL21R−/− mice had greater cerebral infarct volumes performed with VEGFR1 selective ligands in ischemic
when compared with wild-type C57/BL6 mice in a heart or PAD models concluded that the mechanism
stroke model. Furthermore, within that study, a coding for the angiogenic effects of these factors was through
Downloaded from http://ahajournals.org by on July 26, 2023

variant between C57BL/6 and Balb/c mice at amino occupying VEGFR1, thus allowing VEGF165a and other
acid 200, in the Balb/c allele of IL-21R was linked to ligands to preferentially activate VEGFR2.81–86 This is
greater neuronal injury and poorer stroke outcomes. At often referred to as a decoy effect.87
200 AA, humans express the C57BL/6 sequence, with Using a similar approach as Kikuchi et al,77 in Balb/C
only very rare alternative coding variants. Ongoing work mice, we found therapeutic benefit following HLI with
is focused on the IL-21R mediated, nitric oxide-indepen- delivery of the VEGF165b antibody.40 In these in vitro and
dent, angiogenesis in PAD. in vivo studies, we consistently found that VEGF165b
impaired endothelial VEGFR1 (not VEGFR2) activa-
tion.40 Administration of the VEGF165b antibody reduced
Refining Angiogenesis and Modulating binding of VEGF165b to VEGFR1 and increased the
Antiangiogenic Factors activation of VEGFR1, as indicated by its level of phos-
Within the well-studied VEGF receptor-ligand family, phorylation at Y1333, and this occurred with coimmu-
VEGFR2 is widely regarded as the primary receptor noprecipitation with activated STAT3. In this report, we
whose activation drives hypoxia-dependent and post- concluded that when VEGF165b reduces the activation
natal angiogenesis.71 VEGF-A is one of several struc- of VEGFR1, the antibody against VEGF165b augments
turally related ligands for the VEGFR tyrosine kinases: VEGFR1-mediated angiogenesis. This is not meant to
VEGFR1, R2, and R3.71–73 Within the VEGF-A gene, imply that VEGF165b completely silences VEGFR1. Nota-
there are major alternative splicing sites resulting in bly, in wild-type Balb/C and C57/Bl6 mice after HLI,
proteins of varying lengths of 121, 165, and 189 amino both VEGFR1 and VEGF165b are increased, and there-
acids. There is a 206 splice variant whose expression fore, the level ofVEGFR1 activation is dependent upon
is limited to development. The longer transcripts have the levels of VEGF165b and other VEGFR1 ligands that
a stronger capacity to bind to the extracellular matrix, are competing for binding.88,89 This is consistent with
which alters their bioavailability. Strategies are avail- published data that kinase-dead knockout mice and
able to activate multiple isoforms, and these include VEGFR1+/− knockout mice have impaired angiogenesis
activating transcription factors and the use of single and perfusion recovery after HLI when compared with
plasmid encoding multiple transcripts. Rebar et al74 wild type.90 Within macrophages, VEGFR1 activation
reported that the administration of a single isoform promotes an M2-like angiogenic state in macrophages

1948   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

which is enhanced by inhibition of VEGF165b.40 Thus, in shown to be p21, p53, and E2F1, while in macrophages,

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
the setting of PAD, the VEGFR1 pathway is regulated it was immune response factor-9.39,98
differently than VEGFR2 and may activate angiogenic Predicting the net physiological effect of miR modu-
pathways by themselves or ones that can have therapeu- lation in PAD conditions is not always straightforward.
tic synergy with the VEGFR2/eNOS pathway. Although miR-155 has antiangiogenic effects in target-
ing eNOS, and VEGF levels are inversely correlated with
Role of Noncoding RNAs (miR and Long Noncoding miR-155 expression, the inhibition of miR-155 attenu-
RNA) ates blood flow recovery and leukocyte recruitment in
Micro-RNAs are 15 to 23 nucleotide (noncoding) vivo following HLI.107–109 Indeed, the totality of data sug-
RNAs that have emerged as key regulators of the gests a negative role for miR-155 in the response to HLI.
response to hemodynamic forces, vascular injury, and Thus, in considering miRs for therapeutic modulation, rel-
hypoxia.91,92 A miR is thought to primarily exert its evant disease models can be informative. In this regard,
effects by incorporating into the RNA-induced silenc- however, not all miRs, or the gene targets for a given
ing complex which then binds to a target mRNA, usu- miR, are conserved through evolution and miRs that have
ally in the 3′ UTR of its target mRNA.93,94 The binding retained homology and function from rodents to humans
of the miR to the mRNA can suppress a single gene may be preferential for clinical development.
target or the miR can have the capacity to regulate Another class of noncoding RNAs is the long non-
multiple genes within a common pathway simultane- coding RNAs (lncRNAs) which are typically defined
ously by promoting degradation of the mRNA or by as RNAs of >200 nucleotides.110 Reports are begin-
inhibiting mRNA translation to protein.94,95 ning to emerge on the potential role of lncRNAs in
Although a full review of noncoding RNA in angiogen- PAD.111 When compared with miRs, lncRNAs are far
esis and arteriogenesis is beyond the scope of this review, more complex. In general, lncRNAs are known to be
herein, we will provide examples of noncoding RNAs and far more species-specific and they contain numerous
their potential application for future vascular therapeutics potential splice variants. A lncRNA can serve numerous
in PAD. The first reports of miRs that were identified as potential regulatory functions from acting as epigenetic
candidates for modulation of neovascularization in PAD modifiers, to regulating gene expression, to modifying
were of miR-92a96 and miR-100.97 Both of these miRs protein translation, and acting as protein scaffolds.112,113
were identified based upon the response of EC to injury. For example, the lncRNA LEENE facilitates the recruit-
Subsequently, the role of each in PAD-related angiogen- ment of RNA Polymerase II to the eNOS promoter to
Downloaded from http://ahajournals.org by on July 26, 2023

esis was tested in preclinical models where the inhibition enhance eNOS RNA transcription.114
of the miR, by its antagomirs, enhanced perfusion recov- SENCR is a lncRNA that is highly expressed in EC
ery.96,97 Antagomirs (antisense miRs) have been shown and is necessary for EC proliferation, migration, and tube
to have a high degree of specificity and beyond having formation in vitro.115,116 Intriguingly, SENCR is expressed
little affect in a cell or tissue where the target miR is not at lower levels in the vascular tissue of patients with
expressed; antagomers even in target tissue can expres- CLI.116 For reasons that include the potential to exam-
sion levels of closely related miR’s unchanged.98 Studies ine rodent models, lncRNAs that are conserved from
that focused on the ability of miRs to modulate angio- mouse to human are far easier to study than are those
genesis in PAD conditions appeared.98–103 The response, that are not conserved. Human-specific lncRNA cannot
that is, the gene or gene pathway, modulated by a miR readily be used in preclinical models of PAD to examine
can be expected to be different between cells, or target effects on angiogenesis, arteriogenesis, perfusion recov-
tissues, based upon the presence of an injury.103 Moving ery, and potentially tissue loss. Studies have sought to
from angiogenesis to arteriogenesis, Landskroner-Eiger identify candidate lncRNAs based on their association
et al104 focused on miR(s) that regulated arteriogenesis. with angiogenesis and have, for example, characterized
In vitro studies under shear stress, identified candidate the response of ECs to hypoxia in vitro where lncRNAs,
miRs that could regulate arteriogenesis, and in murine including LNC00323, MIR503-HG, GATA-AS, have all
PAD models, approaches that targeted miR-199a and been shown to be differentially regulated in human EC
miR-146a demonstrated improved perfusion recovery with after hypoxic injury.117–120 Further analysis showed
following femoral artery ligation.105,106 Using the same that a targeted knockdown of LINC00323 impaired EC
approach described in the identification of the IL-21R, tube formation while MIR503-HG knockdown reduced
we used unbiased genetic, computational, and experi- EC migration, although both were necessary for prolif-
mental approaches and in vivo HLI models where we eration.117 Voellenkle et al120 found H19, MIR210HG,
examined the target tissue in ischemic skeletal muscle MEG9, MALAT1, and MIR22HG to be elevated by
and identified miR-93 as a potential therapeutic target hypoxia in human umbilical vein ECs in vitro and because
to modulate PAD.98 For miR-93, although overexpres- these were evolutionarily conserved, they were able to be
sion has beneficial effects on ECs and macrophages in examined in mouse gastrocnemius muscle following HLI.
PAD relevant conditions, the target gene(s) for ECs was MALAT1 inhibition impaired angiogenesis in vitro and

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1949


Annex and Cooke Vascular Regeneration in PAD

blood flow recovery and reduced capillary density fol- oxygen tension.68,98 In the state of hypoxia with serum star-
DISEASE COMPENDIUM
PERIPHERAL VASCULAR

lowing HLI in vivo.118 In aged mice, MEG3 inhibition was vation, the addition of VEGF165a to ECs induces angiogen-
shown to improve blood flow recovery following HLI.121 esis by reducing cell death and increasing EC proliferation
The systematic examination of human tissue will likely in association with an increase in glycolysis.133,134 However,
offer the best way to identify lncRNAs that may play a in VEGF-mediated tumor angiogenesis, there is vascular
role in, or serve as therapeutic targets for, PAD.122 permeability and leaky blood vessels.133,134 Whether all forms
of angiogenesis use the same EC metabolism pathways
Endothelial Metabolism in Angiogenesis.
as tumors is not completely understood nor are the poten-
Research in therapeutic angiogenesis, certainly from the
tial consequences of using metabolic pathways other than
cardiovascular perspective, has largely focused on the
glycolysis.133–135
use of cytokine growth factors to bind to and activate
receptors on the EC surface to drive receptor signal- Generating Better Autologous Cell Therapies
ing within in vitro systems to move EC into a prolifera- As described briefly above, large randomized, well-con-
tive state. Even when in a quiescent state, energy from ducted clinical trials of cell therapies for PAD, such as the
ATP is required for ECs to maintain physiological func- National Heart, Lung, and Blood Institute-funded PACE
tion and homeostasis. In ECs, the majority of ATP is pro- study, have not shown benefit of adult stem cell therapy.
duced via glycolytic metabolism, and this phenomenon It is possible that the difference between the preclinical
occurs despite the location of ECs being adjacent to models (where many cell therapies have efficacy), and the
the bloodstream and in contact with the sufficient lev- clinical trials, is related to the number and the quality of the
els of oxygen.123 In ECs, the 6-phosphofructo-2-kinase/ autologous stem cells derived from individuals with cardio-
fructose-2,6-biphosphatase enzymes PFKFBs prod- vascular disease.136 In this regard, the ability to provide suf-
uct is an activator of PFK1 (phosphofructokinase 1), a ficient numbers of high-quality therapeutic cells is likely to
rate-limiting enzyme of glycolysis.124 Although there are be critical in obtaining a reproducible and significant benefit.
4 isoforms of PFKFB, PFKFB3 has a kinase/phospha- In any event, different approaches are needed. One novel
tase activity >700, the kinase to phosphatase activity in approach to cell therapy for PAD is to generate therapeutic
PFKFB2 and PFKFB4 are 1.8 and 0.9, respectively, and cells using autologous skin fibroblasts.137 In this case, a field
the expression of PFKFB1 in ECs is low.124,125 Therefore, of fibroblasts are exposed to retroviral vectors encoding
PFKFB3 is critical for converting F6P to F2,6BP, with the master regulators of pluripotency (eg, Oct 4 [octamer-
F2,6BP being the main allosteric activator of PFK1; the binding transcription factor 4], Sox2 [SRY (sex-determining
Downloaded from http://ahajournals.org by on July 26, 2023

most important regulatory enzymes of glycolysis. Taken region Y)-box 2], KLF4 [Kruppel-like factor 4], and cMyc
together, PFKFB3 is critical for the regulation of endo- [cellular myelocytomatosis]). These transcriptional factors
thelial glycolysis.123–126 direct the activation of a cascade of factors required for
Beyond homeostasis, ECs need ATP for angiogenesis. pluripotency, and over a period of several weeks, many of
Tumor angiogenesis is perhaps the most extensively studied the fibroblasts will become induced pluripotent stem cells
angiogenesis systems and one in which the growth/prolifer- (iPSCs). Subsequently, the iPSCs can be differentiated into
ation of ECs requires an increase in glycolytic metabolism.127 any of the 3 germ layers, and further differentiated into the
Exposing ECs to hypoxia alone has been used to model lineage of choice, using growth factors and small molecules
conditions such as tumor angiogenesis and retinopathy.128 that facilitate the differentiation into a particular lineage,
The changes in EC metabolism observed under hypoxia are such as ECs. Rufaihah et al138 have previously shown that
similar to what is seen with exposure of ECs to VEGF165a, iPSC-derived ECs manifest all of the expected characteris-
where EC growth and proliferation is accompanied with tics and functions, expressing endothelial surface markers
an increase in glycolytic metabolism that occurs with, and (eg, VE-cadherin), generating nitric oxide and angiogenic
is dependent on, an upregulation of PFKFB3 to increase cytokines, and forming vascular networks in Matrigel. Fur-
the glycolytic flux.129–131 In human umbilical vein ECs chal- thermore, when injected into the underperfused limb of the
lenged with hypoxia only, PFKFB3 knockdown using siRNA murine HLI model, perfusion was increased. Interestingly,
or pharmacological inhibition has resulted in decreased cell when delivered systemically the ECs derived from pluripo-
proliferation.124 Tumors which are known to use enhanced tent stem cells can migrate to the ischemic hindlimb and
glycolysis for angiogenesis when embedded in EC-specific increase perfusion as assessed by bioluminescence and
PFKFB3 knockout mice show slower growth rates than laser doppler spectroscopy.139 These findings suggest that
tumors transplanted in control mice.125 Attempts to promote autologous iPSC-derived ECs might be useful as a cell
blood vessel growth in the ischemic muscle bed to improve therapy in patients with PAD. However, iPSC derivatives will
leg blood flow with agents such as VEGF165a has been met need to be rigorously tested for the fidelity of reprogram-
with limited therapeutic success in humans.56,132 The angio- ming and the exclusion of pluripotent cells from the thera-
genesis that occurs in ischemic leg muscle, when compared peutic product.
with angiogenesis in a tumor, is occurring in a tissue that is Another approach to generating autologous ECs is to
relatively low in both cytokine growth factors and, of course, reprogram fibroblasts directly into ECs.140–142 This form of

1950   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

nuclear reprogramming from one somatic cell lineage to a subsequent paper supported by extensive lineage trac-

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
another is also known as direct reprogramming or trans- ing found no evidence of MEndoT in the murine myocar-
differentiation so as to distinguish it from the process dial infarction model.152
where the generation of an iPSC is an intermediate step. The contribution and extent of potential modulation of
Transdifferentiation is a change from one somatic cell MEndoT in the response to limb ischemia is still in the pro-
type to another. In addition, the term transdifferentiation cess of being understood. Because there are significant
may also apply to radical changes in cell form or function differences in peripheral versus myocardial tissues, in their
within a cell lineage, such as when a senescent EC is vascularity and their response to disease, lineage tracing
transformed into a juvenile phenotype, as with the forced and single-cell RNA sequencing were used in an HLI model,
expression of telomerase.143 Whether transdifferentiation to detect evidence of MEndoT. Surprisingly, 8 distinctly dif-
is across somatic lineages, or across time as in the latter ferent subpopulations of fibroblasts in the mouse hindlimb,
example, the cell must overcome epigenetic barriers that confirmed by their high expression of genes known to be
maintain cell phenotype, and must rearrange chromatin preferentially expressed in fibroblasts (eg, Tcf4/Tcf7l2,
configuration so as to support the global change in tran- fibronectin, and collagen IV) yet distinct by their global tran-
scriptional profile that underlies a new cell identity.144,145 scriptional profile. Two subpopulations expressed some EC
In the case of direct reprogramming, retroviral vectors genes at baseline and expanded dramatically during isch-
or mRNA encoding the master regulators of endothe- emia. Genetic or pharmacological suppression of inflamma-
lial identity (eg, ETV2, FLI1 [Friend leukemia integra- tory signaling abrogated the expansion of this population
tion 1 transcription factor], GATA2 [GATA-binding factor and impaired vascularity, recovery of perfusion, and preser-
2], and KLF4) can induce fibroblasts to become ECs. vation of ischemic tissue. Notably, surface markers for these
These induced ECs are similar to iPSC-ECs in endothe- 2 populations permitted their isolation from the ischemic
lial functions, as well as the ability to restore perfusion hindlimb by fluorescence-activated cell sorting. Whereas
when injected into the ischemic limb of the mouse.146 In most fibroblasts will form clusters of cells in Matrigel, one of
either case, the generation of the induced pluripotent the subpopulations reproducibly formed networks and dis-
stem cell or the direct reprogramming of a fibroblast to played surface markers characteristic of ECs when placed
an EC requires the activation of cell-autonomous innate into Matrigel.153
immune signaling.147 The activation of innate immune The expansion or transdifferentiation of resident
signaling increases DNA accessibility to facilitate the fibroblasts to ECs depends upon inflammatory signaling,
action of lineage determination factors to change the cell an observation consistent with our prior work showing
Downloaded from http://ahajournals.org by on July 26, 2023

phenotype.147–149 The process by which inflammatory sig- that inflammatory signaling promotes DNA accessibility
naling increases DNA accessibility to facilitate changes and phenotypic plasticity.148,153 Hypoxic injury (generat-
in cell fate is termed transflammation (Figure 2). ing damage-associated molecular patterns or DAMPs)
Specific caveats regarding the use of reprogrammed and inflammatory cytokines released in the setting of
cells for therapy relates to concerns regarding residual ischemia activate inflammatory signaling (Figure 2). This
pluripotent stem cells that could generate abnormal tis- triggers global changes in epigenetic modifiers (such as
sues (in the case of iPSC-ECs); and the fidelity of the upregulation of histone acetylases and downregulation
reprogramming to functional ECs.150 Other general con- of histone deacetylases) which favor an open chromatin
cerns of cell therapy remain, for example, dosing, duration configuration.154 In addition, activation of iNOS (induc-
of treatment, delivery methods, and cellular persistence ible nitric oxide synthase) facilitates S-nitrosylation
in a hostile ischemic environment and to what extent the of the polycomb repressive complex 1 and NURD1
environment can be modulated before therapy. (nucleosome remodelling and deacetylase 1), causing
the dissociation of these repressive complexes from the
Generating Autologous Therapeutic Cells In Situ chromatin.148,149 Finally, inflammatory signaling induces a
If autologous therapeutic cells could be generated in situ, glycolytic shift, which supplies the nucleus with metabo-
it might solve the issues regarding delivery of the cells lites that participate in epigenetic modification. For exam-
and could potentially reverse processes contributing to ple, the glycolytic shift is associated with citrate export
disease, such as endothelial-to-mesenchyme transition, from the mitochondria to the nucleus, where it is con-
which may contribute to scarring in the setting of isch- verted to acetylcoA for histone acetylation.155
emic injury. Indeed, there is evidence that fibroblast-to- To summarize, single-cell profiling and lineage trac-
endothelial transdifferentiation (MEndoT) contributes to ing has revealed that there may be some contribution of
the recovery from ischemia in vivo, although this concept specific populations of tissue fibroblasts to angiogenesis
remains controversial.137,141,142,147–149 Ubil et al151 have in limb ischemia. It may be possible to enhance cellu-
presented evidence that resident fibroblasts may trans- lar plasticity to enhance therapeutic transdifferentiation
differentiate into ECs, to increase capillary density and of fibroblasts to angiogenic cells. Drugs which favorably
to enhance perfusion in the murine myocardial infarction affect the activity of epigenetic modifiers or the glycolytic
model which is mediated partially through p53. However, state are worthy of further study for PAD.

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1951


DISEASE COMPENDIUM Annex and Cooke Vascular Regeneration in PAD
PERIPHERAL VASCULAR

Figure 2. Damage-associated
molecular patterns (DAMPs)
generated by hypoxic injury
stimulate PRRs (pattern recognition
receptors) which activate signaling
pathways that alter transcription or
posttranslational modifications of
epigenetic enzymes to promote DNA
accessibility.
In addition, a glycolytic shift supplies
metabolites to the nucleus for histone
modifications that further facilitate an
open chromatin configuration. Together
with environmental influences that favor
an endothelial phenotype (eg, VEGF
[vascular endothelial growth factor]),
fibroblasts undergo a transdifferentiation
to angiogenic cells that generate vascular
growth factors and incorporate into the
microvasculature.
Downloaded from http://ahajournals.org by on July 26, 2023

the application of contemporary molecular technologies


EPILOGUE to human samples may be most useful in generating
The past 25 years of research into angiogenic and cell novel therapeutic avenues. For example, genome-wide
therapies for PAD have not yet translated into significant association studies have provided support for antithrom-
changes in PAD management. The discordance between botic treatment of PAD,156 a finding that is consistent
what we have learned and what we have accomplished with the improved limb outcomes in patients with PAD
therapeutically raises important questions. Are the ani- with low dose rivaroxaban in the Compass study. Ongoing
mal models that we are using misdirecting our therapeu- studies using single-cell multiomics (eg, single-cell RNA
tic trials? The classic hindlimb ischemia model in mice is sequencing, genome sequencing, and ATAC sequencing)
typically an acute femoral artery ligation, in the absence applied to human tissues will also provide novel insights
of cardiovascular risk factors such as hypercholester- into disease mechanisms. Such studies may provide use-
olemia, hypertension, tobacco exposure, diabetes, or ful information such as target receptor expression in sub-
senescence. It is critical to consider the effects of risk groups of patients or better validate molecular targets
factors (in particular diabetes and tobacco exposure) in identified in murine models.
preclinical models. Better animal models are available It is critical to remember that in patients with PAD,
(eg, hypercholesterolemic cynomolgus monkeys) but there is substantial patient heterogeneity in the response
are prohibitively expensive and low throughput. A more to similar degrees of atherosclerotic disease. Patients
feasible approach may be to preferentially use genetic with the same hemodynamic compromise may have dif-
mouse models that combine cardiovascular risk factors ferent presentations. Why one patient has IC, whereas
and use modifications to the classic ligation/excision to another has CLI is not entirely explained by the large
more closely mimic human conditions. An animal model vessel disease, suggesting that there are microvascular
that develops peripheral arterial lesions would be ideal or end-organ differences. In which case, the pathophysi-
but those will need to reach a state of hemodynamically ology, and the therapeutic reversal of disease, may be
significant occlusion. Conversely, multiorgan chip models different in these 2 sets of patients.
that combine microfluidics and physiological hemody- Is the failure of angiogenic and cell therapies related
namic forces with human cells could be useful. Finally, to our tendency to do single-agent studies? Would a

1952   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

combinatorial approach be more successful? Rather of angiogenesis, arteriogenesis, adult vasculogenesis,

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
than just facilitating angiogenesis, should we also be and cell fate decisions regulating vascular recovery. The
considering other processes such as endothelial per- answers lie in the refinement of our models, in the appli-
meability and pericyte association, which may translate cation of new molecular tools to patient tissues, and in
into less permeable, more functional, and more stable the persistence of our efforts to benefit our patients.
vessels? What about other processes that may contrib-
ute to angiogenesis, such as clonal EC expansion?157
What is the role of the skeletal muscle itself in vasculo- ARTICLE INFORMATION
genesis? Certainly, metabolic products during exercise Affiliations
affect vasomotion. More studies of how such metabolic Vascular Biology Center, Department of Medicine, Medical College of Georgia,
products effect maintenance or repair of the microvas- Augusta University (B.H.A.). Department of Cardiovascular Sciences, Houston
Methodist Research Institute, TX (J.P.C.).
culature are needed.
Are iPSC-derived cells a better form of cell therapy? Sources of Funding
Can we ensure the fidelity of reprogramming and the sta- Dr Annex is supported by R01HL150003, RO1148590, RO1HL141325,
3RO1HL101200 (Popel, Johns Hopkins, PI), and RO1GM129074 (Mac
bility of cell fate? Will such cells be more resistant to the Gabhann, Johns Hopkins, PI). Dr Cooke is supported by R01s HL133254
hostile environment into which they are placed? And how and HL148338; and Cancer Prevention Research Institute of Texas CPRIT
will we deliver such cells, and in what dose and duration? RP150611.
Is it possibly better to transdifferentiate resident meso- Disclosures
dermal cells into ECs that can expand the microvascula- Dr Annex is the founder of Merand Pharmaceutical a start-up company estab-
ture? Can we restore tissue plasticity and regeneration lished at the University of Virginia to commercialize miR-93 for peripheral arte-
rial disease (PAD) but the work for this article was supported by the National
by epigenetic-directed therapies? Or is any of this going Institutes of Health (NIH) grants to Dr Annex before the formation of the com-
to be useful without first restoring flow through the large pany and no financial or time support from Merand Pharmaceutical was used.
conduit vessels? Dr Cooke is on the Scientific Advisory Boards for Humann, Fibralign, and Ja-
nOne and is the majority owner of Cooke Consulting; none of which companies
We may also need to rethink how we test novel provided financial support for this work. No products of these companies were
PAD therapies in clinical trials. For example, in trials of discussed in this review.
angiogenic agents, hemodynamic, and anatomic assess-
ments, such as low ankle-brachial blood pressure index
and angiography, are poor surrogate measurements for REFERENCES
perfusion. Treadmill studies to assess function may be
Downloaded from http://ahajournals.org by on July 26, 2023

1. Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng
complicated by many factors including the fibro-fatty S, Chiuve SE, Cushman M, Delling FN, Deo R, et al; American Heart Asso-
ciation Council on Epidemiology and Prevention Statistics Committee and
replacement of skeletal muscle that attends chronic limb Stroke Statistics Subcommittee. Heart Disease and Stroke Statistics-2018
ischemia. Thus, improved measures to assess improve- Update: a Report From the American Heart Association. Circulation.
ments in muscle perfusion and function will be advan- 2018;137:e67–e492. doi: 10.1161/CIR.0000000000000558
2. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott
tageous. For cell therapy trials, do we have the proper MM, Norman PE, Sampson UKA, Williams LJ, Mensah GE, et al. Com-
tools, for example, imaging, cell tracking, etc for conduct- parison of global estimates of prevalence and risk factors for peripheral
ing more informative early phase human studies? Inflam- artery disease in 2000 and 2010: a systematic review and analysis. Lancet.
2013;382:1329–40. doi: 10.1016/S0140-6736(13)61249-0
mation plays a major role in coronary artery disease, and 3. Hirsch AT, Duval S. The global pandemic of peripheral artery disease. Lan-
likely plays an important role in PAD. However, emerging cet. 2013;382:1312–1314. doi: 10.1016/S0140-6736(13)61576-7
evidence indicates that inflammatory signaling is impor- 4. Kullo IJ, Rooke TW. CLINICAL PRACTICE. Peripheral artery disease. N Engl
J Med. 2016;374:861–871. doi: 10.1056/NEJMcp1507631
tant for vascular regeneration.158 Methods are needed 5. Allison MA, Ho E, Denenberg JO, Langer RD, Newman AB, Fabsitz RR,
to assess inflammatory signaling in patient tissue, so as Criqui MH. Ethnic-specific prevalence of peripheral arterial disease in
to optimally modulate this process to enhance vascular the United States. Am J Prev Med. 2007;32:328–333. doi: 10.1016/j.
amepre.2006.12.010
anatomy, perfusion, and skeletal muscle function. For 6. Nead KT, Cooke JP, Olin JW, Leeper NJ. Alternative ankle-brachial
patients with CLI, the major criteria for Food and Drug index method identifies additional at-risk individuals. J Am Coll Cardiol.
Administration approval are amputation-free survival. 2013;62:553–559. doi: 10.1016/j.jacc.2013.04.061
7. Selvin E, Erlinger TP. Prevalence of and risk factors for peripheral arterial
However, for these patients, should not pain relief and disease in the United States: results from the National Health and Nutri-
reduction in narcotic use be approvable end points? tion Examination Survey, 1999-2000. Circulation. 2004;110:738–43. doi:
10.1161/01.CIR.0000137913.26087.F0
8. Olin JW, Sealove BA. Peripheral artery disease: current insight into the dis-
CONCLUSIONS ease and its diagnosis and management. Mayo Clin Proc. 2010;85:678–
692. doi: 10.4065/mcp.2010.0133
These are just a few of the difficult questions that face 9. 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
investigators and regulators in the field of vascular Disease after Revascularization. N Engl J Med. 2020;382:1994–2004. doi:
regeneration. However, it is our feeling that the answers 10.1056/NEJMoa2000052
to these questions are within reach. In recent years, there 10. Gerhard-Herman MD, Gornik HL, Barrett C, Barshes NR, Corriere MA,
Drachman DE, Fleisher LA, Fowkes FGR, Hamburg NM, Kinlay S, et al.
have been significant advances in our understanding of 2016 AHA/ACC Guideline on the Management of Patients With Lower
the genetic, epigenetic, proteomic, and cellular regulation Extremity Peripheral Artery Disease: executive Summary: a Report of the

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1953


Annex and Cooke Vascular Regeneration in PAD

American College of Cardiology/American Heart Association Task Force on novel angiogenic protein: its role in ischemia. Circulation. 2004;109:1314–
DISEASE COMPENDIUM
PERIPHERAL VASCULAR

Clinical Practice Guidelines. J Am Coll Cardiol. 2017;69:1465–1508. doi: 1319. doi: 10.1161/01.CIR.0000118465.36018.2D
10.1016/j.jacc.2016.11.008 31. Wolf C, Cai WJ, Vosschulte R, Koltai S, Mousavipour D, Scholz D,
11. Baubeta Fridh E, Andersson M, Thuresson M, Sigvant B, Kragsterman Afsah-Hedjri A, Schaper W, Schaper J. Vascular remodeling and altered
B, Johansson S, Hasvold P, Falkenberg M, Nordanstig J. Amputation protein expression during growth of coronary collateral arteries. J Mol Cell
rates, mortality, and pre-operative comorbidities in patients revascula- Cardiol. 1998;30:2291–2305. doi: 10.1006/jmcc.1998.0790
rised for intermittent claudication or critical limb ischaemia: a popula- 32. Cai WJ, Koltai S, Kocsis E, Scholz D, Kostin S, Luo X, Schaper W, Schaper J.
tion based study. Eur J Vasc Endovasc Surg. 2017;54:480–486. doi: Remodeling of the adventitia during coronary arteriogenesis. Am J Physiol
10.1016/j.ejvs.2017.07.005 Heart Circ Physiol. 2003;284:H31–H40. doi: 10.1152/ajpheart.00478.2002
12. Criqui MH, Aboyans V. Epidemiology of peripheral artery disease. Circ Res. 33. Fung E, Helisch A. Macrophages in collateral arteriogenesis. Front Physiol.
2015;116:1509–1526. doi: 10.1161/CIRCRESAHA.116.303849 2012;3:353. doi: 10.3389/fphys.2012.00353
13. Vartanian SM, Conte MS. Surgical intervention for peripheral arterial disease. 34. Heuslein JL, Meisner JK, Li X, Song J, Vincentelli H, Leiphart RJ, Ames
Circ Res. 2015;116:1614–1628. doi: 10.1161/CIRCRESAHA.116.303504 EG, Blackman BR, Blackman BR, Price RJ. Mechanisms of ampli-
14. McDermott MM, Ades P, Guralnik JM, Dyer A, Ferrucci L, Liu K, Nelson fied arteriogenesis in collateral artery segments exposed to reversed
M, Lloyd-Jones D, Van Horn L, Garside D, et al. Treadmill exercise and flow direction. Arterioscler Thromb Vasc Biol. 2015;35:2354–2365. doi:
resistance training in patients with peripheral arterial disease with and 10.1161/ATVBAHA.115.305775
without intermittent claudication: a randomized controlled trial. JAMA. 35. Heil M, Ziegelhoeffer T, Wagner S, Fernández B, Helisch A, Martin S, Tribulova
2009;301:165–74. doi: 10.1001/jama.2008.962. Erratum in: JAMA. S, Kuziel WA, Bachmann G, Schaper W. Collateral artery growth (arterio-
2012;307:1694. genesis) after experimental arterial occlusion is impaired in mice lacking
15. Garg PK, Liu K, Tian L, Guralnik JM, Ferrucci L, Criqui MH, Tan J, CC-chemokine receptor-2. Circ Res. 2004;94:671–7. doi: 10.1161/01.
McDermott MM. Physical activity during daily life and functional decline RES.0000122041.73808.B5
in peripheral arterial disease. Circulation. 2009;119:251–260. doi: 36. Krishnasamy K, Limbourg A, Kapanadze T, Gamrekelashvili J, Beger C,
10.1161/CIRCULATIONAHA.108.791491 Häger C, Lozanovski VJ, Falk CS, Napp LC, Bauersachs J, et al. Blood ves-
16. Annex BH. Therapeutic angiogenesis for critical limb ischaemia. Nat Rev sel control of macrophage maturation promotes arteriogenesis in ischemia.
Cardiol. 2013;10:387–396. doi: 10.1038/nrcardio.2013.70 Nat Commun. 2017;8:952. doi: 10.1038/s41467-017-00953-2
17. Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangio- 37. Zbinden S, Zbinden R, Meier P, Windecker S, Seiler C. Safety and effi-
genesis. Nat Rev Mol Cell Biol. 2007;8:464–478. doi: 10.1038/nrm2183 cacy of subcutaneous-only granulocyte-macrophage colony-stimulating
18. Mentzer SJ, Konerding MA. Intussusceptive angiogenesis: expansion and factor for collateral growth promotion in patients with coronary artery
remodeling of microvascular networks. Angiogenesis. 2014;17:499–509. disease. J Am Coll Cardiol. 2005;46:1636–1642. doi: 10.1016/j.
doi: 10.1007/s10456-014-9428-3 jacc.2005.01.068
19. Aicher A, Rentsch M, Sasaki K, Ellwart JW, Fändrich F, Siebert R, Cooke JP, 38. Caradu C, Couffinhal T, Chapouly C, Guimbal S, Hollier PL, Ducasse E,
Dimmeler S, Heeschen C. Nonbone marrow-derived circulating progenitor Bura-Rivière A, Dubois M, Gadeau AP, Renault MA. Restoring endothe-
cells contribute to postnatal neovascularization following tissue ischemia. Circ lial function by targeting desert Hedgehog downstream of Klf2 improves
Res. 2007;100:581–589. doi: 10.1161/01.RES.0000259562.63718.35 critical limb Ischemia in adults. Circ Res. 2018;123:1053–1065. doi:
20. Rehman J, Li J, Parvathaneni L, Karlsson G, Panchal VR, Temm CJ, 10.1161/CIRCRESAHA.118.313177
Mahenthiran J, March KL. Exercise acutely increases circulating endothelial 39. Ganta VC, Choi MH, Kutateladze A, Fox TE, Farber CR, Annex BH. A
progenitor cells and monocyte-/macrophage-derived angiogenic cells. J Am MicroRNA93-interferon regulatory factor-9-immunoresponsive gene-
Coll Cardiol. 2004;43:2314–2318. doi: 10.1016/j.jacc.2004.02.049 1-itaconic acid pathway modulates M2-like macrophage polarization to
Downloaded from http://ahajournals.org by on July 26, 2023

21. Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial pro- revascularize Ischemic muscle. Circulation. 2017;135:2403–2425. doi:
genitor cells” are derived from monocyte/macrophages and secrete angio- 10.1161/CIRCULATIONAHA.116.025490
genic growth factors. Circulation. 2003;107:1164–1169. doi: 10.1161/01. 40. Ganta VC, Choi M, Farber CR, Annex BH. Antiangiogenic VEGF165b
cir.0000058702.69484.a0 regulates macrophage polarization via S100A8/S100A9 in periph-
22. Heeschen C, Jang JJ, Weis M, Pathak A, Kaji S, Hu RS, Tsao PS, Johnson eral artery disease. Circulation. 2019;139:226–242. doi: 10.1161/
FL, Cooke JP. Nicotine stimulates angiogenesis and promotes tumor growth CIRCULATIONAHA.118.034165
and atherosclerosis. Nat Med. 2001;7:833–839. doi: 10.1038/89961 41. Libby P. Inflammation in atherosclerosis-No Longer a theory. Clin Chem.
23. Kiuchi K, Matsuoka M, Wu JC, Lima e Silva R, Kengatharan M, Verghese M, 2021;67:131–142. doi: 10.1093/clinchem/hvaa275
Ueno S, Yokoi K, Khu NH, Cooke JP, et al. Mecamylamine suppresses Basal 42. Meier P, Gloekler S, Oezdemir B, Indermuehle A, Traupe T, Vogel R, de Marchi
and nicotine-stimulated choroidal neovascularization. Invest Ophthalmol Vis S, Seiler C. G-CSF induced arteriogenesis in humans: molecular insights
Sci. 2008;49:1705–1711. doi: 10.1167/iovs.07-0089 into a randomized controlled trial. Curr Vasc Pharmacol. 2013;11:38–46.
24. Semenza GL. Oxygen sensing, hypoxia-inducible factors, and disease 43. Grundmann S, Hoefer I, Ulusans S, Bode C, Oesterle S, Tijssen JG, Piek
pathophysiology. Annu Rev Pathol. 2014;9:47–71. doi: 10.1146/annurev- JJ, Buschmann I, van Royen N. Granulocyte-macrophage colony-stimulating
pathol-012513-104720 factor stimulates arteriogenesis in a pig model of peripheral artery disease
25. Qian HS, Liu P, Huw LY, Orme A, Halks-Miller M, Hill SM, Jin F, Kretschmer P, using clinically applicable infusion pumps. J Vasc Surg. 2006;43:1263–
Blasko E, Cashion L, et al. Effective treatment of vascular endothelial growth 1269. doi: 10.1016/j.jvs.2006.02.049
factor refractory hindlimb ischemia by a mutant endothelial nitric oxide syn- 44. Meier P, Gloekler S, de Marchi SF, Indermuehle A, Rutz T, Traupe T, Steck
thase gene. Gene Ther. 2006;13:1342–1350. doi: 10.1038/sj.gt.3302781 H, Vogel R, Seiler C. Myocardial salvage through coronary collateral growth
26. Ferraro B, Cruz YL, Baldwin M, Coppola D, Heller R. Increased perfusion by granulocyte colony-stimulating factor in chronic coronary artery dis-
and angiogenesis in a hindlimb ischemia model with plasmid FGF-2 deliv- ease: a controlled randomized trial. Circulation. 2009;120:1355–1363. doi:
ered by noninvasive electroporation. Gene Ther. 2010;17:763–769. doi: 10.1161/CIRCULATIONAHA.109.866269
10.1038/gt.2010.43 45. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler
27. Morishita R, Nakamura S, Hayashi S, Taniyama Y, Moriguchi A, Nagano T, B, Schatteman G, Isner JM. Isolation of putative progenitor endothe-
Taiji M, Noguchi H, Takeshita S, Matsumoto K, et al. Therapeutic angiogen- lial cells for angiogenesis. Science. 1997;275:964–967. doi: 10.1126/
esis induced by human recombinant hepatocyte growth factor in rabbit science.275.5302.964
hind limb ischemia model as cytokine supplement therapy. Hypertension. 46. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman
1999;33:1379–1384. doi: 10.1161/01.hyp.33.6.1379 ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell traffick-
28. Banfi A, von Degenfeld G, Gianni-Barrera R, Reginato S, Merchant MJ, ing is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat
McDonald DM, Blau HM. Therapeutic angiogenesis due to balanced single- Med. 2004;10:858–864. doi: 10.1038/nm1075
vector delivery of VEGF and PDGF-BB. FASEB J. 2012;26:2486–2497. 47. Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Jung S, Yung S,
doi: 10.1096/fj.11-197400 Chimenti S, Landsman L, Abramovitch R, et al. VEGF-induced adult neo-
29. Lekas M, Lekas P, Mei SH, Deng Y, Dumont DJ, Stewart DJ. Tie2-depen- vascularization: recruitment, retention, and role of accessory cells. Cell.
dent neovascularization of the ischemic hindlimb is mediated by angiopoi- 2006;124:175–189. doi: 10.1016/j.cell.2005.10.036
etin-2. PLoS One. 2012;7:e43568. doi: 10.1371/journal.pone.0043568 48. Yoon CH, Hur J, Park KW, Kim JH, Lee CS, Oh IY, Kim TY, Cho HJ, Kang
30. Ho HK, Jang JJ, Kaji S, Spektor G, Fong A, Yang P, Hu BS, Schatzman HJ, Chae IH, et al. Synergistic neovascularization by mixed transplantation
R, Quertermous T, Cooke JP. Developmental endothelial locus-1 (Del-1), a of early endothelial progenitor cells and late outgrowth endothelial cells:

1954   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

the role of angiogenic cytokines and matrix metalloproteinases. Circulation. nerve stimulation in skeletal muscle. Am J Physiol. 1998;274:H860–H867.

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
2005;112:1618–1627. doi: 10.1161/CIRCULATIONAHA.104.503433 doi: 10.1152/ajpheart.1998.274.3.H860
49. Yoder MC. Defining human endothelial progenitor cells. J Thromb Haemost. 69. Wang T, Cunningham A, Dokun A, Hazarika S, Chen L, Lye R, Leonard W,
2009;7(suppl 1):49–52. doi: 10.1111/j.1538-7836.2009.03407.x Annex B. Angiogenic properties of interleukin-21 under hypoxic conditions.
50. van Royen N, Piek JJ, Buschmann I, Hoefer I, Voskuil M, Schaper W. Blood. 2014: (e-letter to Castermans et al.)
Stimulation of arteriogenesis; a new concept for the treatment of arterial 70. Lee HK, Keum S, Sheng H, Warner DS, Lo DC, Marchuk DA. Natural allelic
occlusive disease. Cardiovasc Res. 2001;49:543–553. doi: 10.1016/s0008- variation of the IL-21 receptor modulates ischemic stroke infarct volume. J
6363(00)00206-6 Clin Invest. 2016;126:2827–2838. doi: 10.1172/JCI84491
51. Heil M, Eitenmüller I, Schmitz-Rixen T, Schaper W. Arteriogenesis versus 71. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor sig-
angiogenesis: similarities and differences. J Cell Mol Med. 2006;10:45–55. nalling - in control of vascular function. Nat Rev Mol Cell Biol. 2006;7:359–
doi: 10.1111/j.1582-4934.2006.tb00290.x 371. doi: 10.1038/nrm1911
52. Robbins JL, Jones WS, Duscha BD, Allen JD, Kraus WE, Regensteiner 72. Cébe-Suarez S, Zehnder-Fjällman A, Ballmer-Hofer K. The role of VEGF
JG, Hiatt WR, Annex BH. Relationship between leg muscle capillary den- receptors in angiogenesis; complex partnerships. Cell Mol Life Sci.
sity and peak hyperemic blood flow with endurance capacity in peripheral 2006;63:601–615. doi: 10.1007/s00018-005-5426-3
artery disease. J Appl Physiol (1985). 2011;111:81–6. doi: 10.1152/ 73. Shibuya M. Differential roles of vascular endothelial growth factor recep-
japplphysiol.00141.2011 tor-1 and receptor-2 in angiogenesis. J Biochem Mol Biol. 2006;39:469–
53. Duscha BD, Robbins JL, Jones WS, Kraus WE, Lye RJ, Sanders JM, Allen 478. doi: 10.5483/bmbrep.2006.39.5.469
JD, Regensteiner JG, Hiatt WR, Annex BH. Angiogenesis in skeletal 74. Rebar EJ, Huang Y, Hickey R, Nath AK, Meoli D, Nath S, Chen B, Xu L,
muscle precede improvements in peak oxygen uptake in peripheral artery Liang Y, Jamieson AC, et al. Induction of angiogenesis in a mouse model
disease patients. Arterioscler Thromb Vasc Biol. 2011;31:2742–2748. doi: using engineered transcription factors. Nat Med. 2002;8:1427–1432. doi:
10.1161/ATVBAHA.111.230441 10.1038/nm1202-795
54. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L, Asahara T, Rosenfield 75. Dai Q, Huang J, Klitzman B, Dong C, Goldschmidt-Clermont PJ,
K, Razvi S, Walsh K, Symes JF. Clinical evidence of angiogenesis after arte- March KL, Rokovich J, Johnstone B, Rebar EJ, Spratt SK, et al. Engi-
rial gene transfer of phVEGF165 in patient with ischaemic limb. Lancet. neered zinc finger-activating vascular endothelial growth factor tran-
1996;348:370–374. doi: 10.1016/s0140-6736(96)03361-2 scription factor plasmid DNA induces therapeutic angiogenesis in
55. Cooke JP, Losordo DW. Modulating the vascular response to limb isch- rabbits with hindlimb ischemia. Circulation. 2004;110:2467–2475. doi:
emia: angiogenic and cell therapies. Circ Res. 2015;116:1561–1578. doi: 10.1161/01.CIR.0000145139.53840.49
10.1161/CIRCRESAHA.115.303565 76. Oltean S, Bates DO. Hallmarks of alternative splicing in cancer. Oncogene.
56. Iyer SR, Annex BH. Therapeutic angiogenesis for peripheral artery dis- 2014;33:5311–5318. doi: 10.1038/onc.2013.533
ease: lessons learned in translational science. JACC Basic Transl Sci. 77. Kikuchi R, Nakamura K, MacLauchlan S, Ngo DT, Shimizu I, Fuster JJ,
2017;2:503–512. doi: 10.1016/j.jacbts.2017.07.012 Katanasaka Y, Yoshida S, Qiu Y, Yamaguchi TP, et al. An antiangiogenic iso-
57. Cooke JP, Meng S. Vascular regeneration in peripheral artery disease. Arte- form of VEGF-A contributes to impaired vascularization in peripheral artery
rioscler Thromb Vasc Biol. 2020;40:1627–1634. doi: 10.1161/ATVBAHA. disease. Nat Med. 2014;20:1464–1471. doi: 10.1038/nm.3703
120.312862 78. Harper SJ, Bates DO. VEGF-A splicing: the key to anti-angiogenic thera-
58. Schleicher M, Yu J, Murata T, Derakhshan B, Atochin D, Qian L, Kashiwagi peutics? Nat Rev Cancer. 2008;8:880–887. doi: 10.1038/nrc2505
S, Di Lorenzo A, Harrison KD, Huang PL, et al. The Akt1-eNOS axis illus- 79. Roberts DM, Kearney JB, Johnson JH, Rosenberg MP, Kumar R,
trates the specificity of kinase-substrate relationships in vivo. Sci Signal. Bautch VL. The vascular endothelial growth factor (VEGF) recep-
2009;2:ra41. doi: 10.1126/scisignal.2000343 tor Flt-1 (VEGFR-1) modulates Flk-1 (VEGFR-2) signaling dur-
Downloaded from http://ahajournals.org by on July 26, 2023

59. Ackah E, Yu J, Zoellner S, Iwakiri Y, Skurk C, Shibata R, Ouchi N, Easton ing blood vessel formation. Am J Pathol. 2004;164:1531–1535. doi:
RM, Galasso G, Birnbaum MJ, et al. Akt1/protein kinase B alpha is 10.1016/S0002-9440(10)63711-X
critical for ischemic and VEGF-mediated angiogenesis. J Clin Invest. 80. Shibuya M, Claesson-Welsh L. Signal transduction by VEGF recep-
2005;115:2119–2127. tors in regulation of angiogenesis and lymphangiogenesis. Exp Cell Res.
60. Murohara T, Asahara T, Silver M, Bauters C, Masuda H, Kalka C, Kearney 2006;312:549–560. doi: 10.1016/j.yexcr.2005.11.012
M, Chen D, Symes JF, Fishman MC, et al. Nitric oxide synthase modulates 81. Silvestre JS, Tamarat R, Ebrahimian TG, Le-Roux A, Clergue M, Emmanuel
angiogenesis in response to tissue ischemia. J Clin Invest. 1998;101:2567– F, Duriez M, Schwartz B, Branellec D, Lévy BI. Vascular endothelial growth
2578. doi: 10.1172/JCI1560 factor-B promotes in vivo angiogenesis. Circ Res. 2003;93:114–123. doi:
61. Dokun AO, Keum S, Hazarika S, Li Y, Lamonte GM, Wheeler F, 10.1161/01.RES.0000081594.21764.44
Marchuk DA, Annex BH. A quantitative trait locus (LSq-1) on mouse 82. Luttun A, Tjwa M, Carmeliet P. Placental growth factor (PlGF) and its receptor
chromosome 7 is linked to the absence of tissue loss after surgical Flt-1 (VEGFR-1): novel therapeutic targets for angiogenic disorders. Ann N
hindlimb ischemia. Circulation. 2008;117:1207–1215. doi: 10.1161/ Y Acad Sci. 2002;979:80–93. doi: 10.1111/j.1749-6632.2002.tb04870.x
CIRCULATIONAHA.107.736447 83. Luttun A, Tjwa M, Moons L, Wu Y, Angelillo-Scherrer A, Liao F, Nagy
62. Wang T, Cunningham A, Dokun AO, Hazarika S, Houston K, Chen L, Lye JA, Hooper A, Priller J, De Klerck B, et al. Revascularization of isch-
RJ, Spolski R, Leonard WJ, Annex BH. Loss of interleukin-21 recep- emic tissues by PlGF treatment, and inhibition of tumor angiogenesis,
tor activation in hypoxic endothelial cells impairs perfusion recovery after arthritis and atherosclerosis by anti-Flt1. Nat Med. 2002;8:831–840.
hindlimb ischemia. Arterioscler Thromb Vasc Biol. 2015;35:1218–1225. doi: doi: 10.1038/nm731
10.1161/ATVBAHA.115.305476 84. Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol M, Wu
63. Wang T, Cunningham A, Houston K, Sharma AM, Chen L, Dokun AO, Lye Y, Bono F, Devy L, Beck H, et al. Synergism between vascular endothelial
RJ, Spolski R, Leonard WJ, Annex BH. Endothelial interleukin-21 receptor growth factor and placental growth factor contributes to angiogenesis and
up-regulation in peripheral artery disease. Vasc Med. 2016;21:99–104. doi: plasma extravasation in pathological conditions. Nat Med. 2001;7:575–
10.1177/1358863X15621798 583. doi: 10.1038/87904
64. Leonard WJ. The Yin and Yang of Interleukin-21 in allergy, autoimmunity 85. Bry M, Kivelä R, Holopainen T, Anisimov A, Tammela T, Soronen J, Silvola J,
and cancer. Blood. 2011;118:1809–1810. Saraste A, Jeltsch M, Korpisalo P, et al. Vascular endothelial growth factor-B
65. Spolski R, Leonard WJ. Interleukin-21: a double-edged sword with thera- acts as a coronary growth factor in transgenic rats without inducing angio-
peutic potential. Nat Rev Drug Discov. 2014;13:381–393. genesis, vascular leak, or inflammation. Circulation. 2010;122:1725–1733.
66. Castermans K, Tabruyn SP, Zeng R, van Beijnum JR, Eppolito C, Leonard doi: 10.1161/CIRCULATIONAHA.110.957332
WJ, Shrikant PA, Griffioen AW. Angiostatic activity of the antitumor cyto- 86. Lähteenvuo JE, Lähteenvuo MT, Kivelä A, Rosenlew C, Falkevall A, Klar J,
kine interleukin-21. Blood. 2008;112:4940–4947. doi: 10.1182/blood- Heikura T, Rissanen TT, Vähäkangas E, Korpisalo P, et al. Vascular endothe-
2007-09-113878 lial growth factor-B induces myocardium-specific angiogenesis and arterio-
67. Palmer-Kazen U, Wariaro D, Luo F, Wahlberg E. Vascular endothelial cell genesis via vascular endothelial growth factor receptor-1- and neuropilin
growth factor and fibroblast growth factor 2 expression in patients with receptor-1-dependent mechanisms. Circulation. 2009;119:845–856. doi:
critical limb ischemia. J Vasc Surg. 2004;39:621–628. doi: 10.1016/j. 10.1161/CIRCULATIONAHA.108.816454
jvs.2003.07.006 87. Boucher JM, Clark RP, Chong DC, Citrin KM, Wylie LA, Bautch VL. Dynamic
68. Annex BH, Torgan CE, Lin P, Taylor DA, Thompson MA, Peters KG, Kraus alterations in decoy VEGF receptor-1 stability regulate angiogenesis. Nat
WE. Induction and maintenance of increased VEGF protein by chronic motor Commun. 2017;8:15699. doi: 10.1038/ncomms15699

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1955


Annex and Cooke Vascular Regeneration in PAD

88. Chu LH, Ganta VC, Choi MH, Chen G, Finley SD, Annex BH, Popel AS. 108. Liu Q, Yang J. Expression and significance of miR155 and vascular endo-
DISEASE COMPENDIUM
PERIPHERAL VASCULAR

A multiscale computational model predicts distribution of anti-angiogenic thelial growth factor in placenta of rats with preeclampsia. Int J Clin Exp
isoform VEGF165b in peripheral arterial disease in human and mouse. Sci Med. 2015;8:15731–7.
Rep. 2016;6:37030. doi: 10.1038/srep37030 109. Pankratz F, Bemtgen X, Zeiser R, Leonhardt F, Kreuzaler S, Hilgendorf
89. Clegg LE, Ganta VC, Annex BH, Mac Gabhann F. Systems Pharmacology I, Smolka C, Helbing T, Hoefer I, Esser JS, et al. MicroRNA-155 exerts
of VEGF165b in peripheral artery disease. CPT Pharmacometrics Syst cell-specific antiangiogenic but proarteriogenic effects during adaptive
Pharmacol. 2017;6:833–844. doi: 10.1002/psp4.12261 neovascularization. Circulation. 2015;131:1575–1589. doi: 10.1161/
90. Amano H, Kato S, Ito Y, Eshima K, Ogawa F, Takahashi R, Sekiguchi K, CIRCULATIONAHA.114.014579
Tamaki H, Sakagami H, Shibuya M, et al. The role of vascular endothelial 110. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding
growth factor receptor-1 signaling in the recovery from Ischemia. PLoS RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22:96–118.
One. 2015;10:e0131445. doi: 10.1371/journal.pone.0131445 doi: 10.1038/s41580-020-00315-9
91. Neth P, Nazari-Jahantigh M, Schober A, Weber C. MicroRNAs in flow- 111. Monteiro JP, Bennett M, Rodor J, Caudrillier A, Ulitsky I, Baker AH.
dependent vascular remodelling. Cardiovasc Res. 2013;99:294–303. doi: Endothelial function and dysfunction in the cardiovascular system: the long
10.1093/cvr/cvt096 non-coding road. Cardiovasc Res. 2019;115:1692–1704. doi: 10.1093/
92. Urbich C, Kuehbacher A, Dimmeler S. Role of microRNAs in vascular dis- cvr/cvz154
eases, inflammation, and angiogenesis. Cardiovasc Res. 2008;79:581– 112. Yu B, Wang S. Angio-LncRs: LncRNAs that regulate angiogenesis and
588. doi: 10.1093/cvr/cvn156 vascular disease. Theranostics. 2018;8:3654–3675. doi: 10.7150/
93. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. thno.26024
2014;15:509–524. doi: 10.1038/nrm3838 113. Hung J, Miscianinov V, Sluimer JC, Newby DE, Baker AH. Targeting non-
94. Jonas S, Izaurralde E. Towards a molecular understanding of microR- coding RNA in vascular biology and disease. Front Physiol. 2018;9:1655.
NA-mediated gene silencing. Nat Rev Genet. 2015;16:421–433. doi: doi: 10.3389/fphys.2018.01655
10.1038/nrg3965 114. Miao Y, Ajami NE, Huang TS, Lin FM, Lou CH, Wang YT, Li S, Kang J,
95. van Rooij E. The art of microRNA research. Circ Res. 2011;108:219–234. Munkacsi H, Maurya MR, et al. Enhancer-associated long non-coding RNA
doi: 10.1161/CIRCRESAHA.110.227496 LEENE regulates endothelial nitric oxide synthase and endothelial func-
96. Bonauer A, Carmona G, Iwasaki M, Mione M, Koyanagi M, Fischer A, tion. Nat Commun. 2018;9:292. doi: 10.1038/s41467-017-02113-y
Burchfield J, Fox H, Doebele C, Ohtani K, et al. MicroRNA-92a controls 115. Lyu Q, Xu S, Lyu Y, Choi M, Christie CK, Slivano OJ, Rahman A, Jin ZG,
angiogenesis and functional recovery of ischemic tissues in mice. Science. Long X, Xu Y, et al. SENCR stabilizes vascular endothelial cell adherens
2009;324:1710–1713. doi: 10.1126/science.1174381 junctions through interaction with CKAP4. Proc Natl Acad Sci U S A.
97. Grundmann S, Hans FP, Kinniry S, Heinke J, Helbing T, Bluhm F, Sluijter JP, 2019;116:546–555. doi: 10.1073/pnas.1810729116
Hoefer I, Pasterkamp G, Bode C, et al. MicroRNA-100 regulates neovascu- 116. Boulberdaa M, Scott E, Ballantyne M, Garcia R, Descamps B, Angelini GD,
larization by suppression of mammalian target of rapamycin in endothelial Brittan M, Hunter A, McBride M, McClure J, et al. A role for the long non-
and vascular smooth muscle cells. Circulation. 2011;123:999–1009. doi: coding RNA SENCR in commitment and function of endothelial cells. Mol
10.1161/CIRCULATIONAHA.110.000323 Ther. 2016;24:978–990. doi: 10.1038/mt.2016.41
98. Hazarika S, Farber CR, Dokun AO, Pitsillides AN, Wang T, Lye RJ, Annex BH. 117. Fiedler J, Breckwoldt K, Remmele CW, Hartmann D, Dittrich M, Pfanne
MicroRNA-93 controls perfusion recovery after hindlimb ischemia by mod- A, Just A, Xiao K, Kunz M, Müller T, et al. Development of long noncod-
ulating expression of multiple genes in the cell cycle pathway. Circulation. ing RNA-based strategies to modulate tissue vascularization. J Am Coll
2013;127:1818–1828. doi: 10.1161/CIRCULATIONAHA.112.000860 Cardiol. 2015;66:2005–2015. doi: 10.1016/j.jacc.2015.07.081
99. Caporali A, Meloni M, Völlenkle C, Bonci D, Sala-Newby GB, Addis R, 118. Michalik KM, You X, Manavski Y, Doddaballapur A, Zörnig M, Braun T,
Downloaded from http://ahajournals.org by on July 26, 2023

Spinetti G, Losa S, Masson R, Baker AH, et al. Deregulation of microR- John D, Ponomareva Y, Chen W, Uchida S, et al. Long noncoding RNA
NA-503 contributes to diabetes mellitus-induced impairment of endothe- MALAT1 regulates endothelial cell function and vessel growth. Circ Res.
lial function and reparative angiogenesis after limb ischemia. Circulation. 2014;114:1389–1397. doi: 10.1161/CIRCRESAHA.114.303265
2011;123:282–291. doi: 10.1161/CIRCULATIONAHA.110.952325 119. Neumann P, Jaé N, Knau A, Glaser SF, Fouani Y, Rossbach O, Krüger M,
100. Yin KJ, Olsen K, Hamblin M, Zhang J, Schwendeman SP, Chen YE. John D, Bindereif A, Grote P, et al. The lncRNA GATA6-AS epigenetically
Vascular endothelial cell-specific microRNA-15a inhibits angiogen- regulates endothelial gene expression via interaction with LOXL2. Nat
esis in hindlimb ischemia. J Biol Chem. 2012;287:27055–27064. doi: Commun. 2018;9:237. doi: 10.1038/s41467-017-02431-1
10.1074/jbc.M112.364414 120. Voellenkle C, Garcia-Manteiga JM, Pedrotti S, Perfetti A, De Toma I,
101. Welten SM, Bastiaansen AJ, de Jong RC, de Vries MR, Peters EA, Boonstra Da Silva D, Maimone B, Greco S, Fasanaro P, Creo P, et al. Implication
MC, Sheikh SP, La Monica N, Kandimalla ER, Quax PH, et al. Inhibition of of Long noncoding RNAs in the endothelial cell response to hy-
14q32 MicroRNAs miR-329, miR-487b, miR-494, and miR-495 increas- poxia revealed by RNA-sequencing. Sci Rep. 2016;6:24141. doi:
es neovascularization and blood flow recovery after ischemia. Circ Res. 10.1038/srep24141
2014;115:696–708. doi: 10.1161/CIRCRESAHA.114.304747 121. Boon RA, Hofmann P, Michalik KM, Lozano-Vidal N, Berghäuser D,
102. Icli B, Li H, Pérez-Cremades D, Wu W, Ozdemir D, Haemmig S, Guimaraes Fischer A, Knau A, Jaé N, Schürmann C, Dimmeler S. Long noncoding
RB, Manica A, Marchini JF, Orgill DP, et al. MiR-4674 regulates angiogenesis RNA Meg3 controls endothelial cell aging and function: implications for
in tissue injury by targeting p38K signaling in endothelial cells. Am J Physiol regenerative angiogenesis. J Am Coll Cardiol. 2016;68:2589–2591. doi:
Cell Physiol. 2020;318:C524–C535. doi: 10.1152/ajpcell.00542.2019 10.1016/j.jacc.2016.09.949
103. Feinberg MW. Healing the injured vessel wall using microRNA-facilitated 122. Terwilliger ZS, Ryan TE, Goldberg EJ, Schmidt CA, Yamaguchi DJ, Karnekar
gene delivery. J Clin Invest. 2014;124:3694–3697. doi: 10.1172/JCI77509 R, Brophy P, Green TD, Zeczycki TN, Mac Gabhann F, et al. Racial dif-
104. Landskroner-Eiger S, Qiu C, Perrotta P, Siragusa M, Lee MY, Ulrich V, ferences in the limb skeletal muscle transcriptional programs of patients
Luciano AK, Zhuang ZW, Corti F, Simons M, et al. Endothelial miR-17~92 with critical limb ischemia. Vasc Med. 2021;6:1358863X20983918. doi:
cluster negatively regulates arteriogenesis via miRNA-19 repression of 10.1177/1358863X20983918. Epub ahead of print.
WNT signaling. Proc Natl Acad Sci U S A. 2015;112:12812–12817. doi: 123. De Bock K, Georgiadou M, Schoors S, Kuchnio A, Wong BW, Cantelmo
10.1073/pnas.1507094112 AR, Quaegebeur A, Ghesquière B, Cauwenberghs S, Eelen G, et al. Role of
105. Heuslein JL, McDonnell SP, Song J, Annex BH, Price RJ. MicroRNA-146a PFKFB3-driven glycolysis in vessel sprouting. Cell. 2013;154:651–663.
regulates perfusion recovery in response to arterial occlusion via arteriogen- doi: 10.1016/j.cell.2013.06.037
esis. Front Bioeng Biotechnol. 2018;6:1. doi: 10.3389/fbioe.2018.00001 124. Ros S, Schulze A. Balancing glycolytic flux: the role of 6-phosphofructo-
106. Heuslein JL, Gorick CM, McDonnell SP, Song J, Annex BH, Price RJ. 2-kinase/fructose 2,6-bisphosphatases in cancer metabolism. Cancer
Exposure of endothelium to biomimetic flow waveforms yields identifi- Metab. 2013;1:8. doi: 10.1186/2049-3002-1-8
cation of miR-199a-5p as a potent regulator of arteriogenesis. Mol Ther 125. Xu Y, An X, Guo X, Habtetsion TG, Wang Y, Xu X, Kandala S, Li Q, Li
Nucleic Acids. 2018;12:829–844. doi: 10.1016/j.omtn.2018.08.001 H, Zhang C, et al. Endothelial PFKFB3 plays a critical role in angiogen-
107. Sun HX, Zeng DY, Li RT, Pang RP, Yang H, Hu YL, Zhang Q, Jiang Y, esis. Arterioscler Thromb Vasc Biol. 2014;34:1231–9. doi: 10.1161/
Huang LY, Tang YB, et al. Essential role of microRNA-155 in regulat- ATVBAHA.113.303041
ing endothelium-dependent vasorelaxation by targeting endothelial ni- 126. Doddaballapur A, Michalik KM, Manavski Y, Lucas T, Houtkooper RH, You
tric oxide synthase. Hypertension. 2012;60:1407–1414. doi: 10.1161/ X, Chen W, Zeiher AM, Potente M, Dimmeler S, et al. Laminar shear stress
HYPERTENSIONAHA.112.197301 inhibits endothelial cell metabolism via KLF2-mediated repression of

1956   June 11, 2021 Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266


Annex and Cooke Vascular Regeneration in PAD

PFKFB3. Arterioscler Thromb Vasc Biol. 2015;35:137–145. doi: 10.1161/ endothelial cells using ER71/ETV2. Circ Res. 2017;120:848–861. doi:

DISEASE COMPENDIUM
PERIPHERAL VASCULAR
ATVBAHA.114.304277 10.1161/CIRCRESAHA.116.309833
127. Zecchin A, Kalucka J, Dubois C, Carmeliet P. How endothelial cells adapt 142. Li Y, Zhou G, Bruno IG, Cooke JP. Telomerase mRNA reverses senes-
their metabolism to form vessels in tumors. Front Immunol. 2017;8:1750. cence in progeria cells. J Am Coll Cardiol. 2017;70:804–805. doi:
doi: 10.3389/fimmu.2017.01750 10.1016/j.jacc.2017.06.017
128. Wong BW, Marsch E, Treps L, Baes M, Carmeliet P. Endothelial cell metab- 143. Matsushita H, Chang E, Glassford AJ, Cooke JP, Chiu CP, Tsao PS.
olism in health and disease: impact of hypoxia. EMBO J. 2017;36:2187– eNOS activity is reduced in senescent human endothelial cells: pres-
2203. doi: 10.15252/embj.201696150 ervation by hTERT immortalization. Circ Res. 2001;89:793–798. doi:
129. Cao Y, Zhang X, Wang L, Yang Q, Ma Q, Xu J, Wang J, Kovacs L, Ayon RJ, 10.1161/hh2101.098443
Liu Z, et al. PFKFB3-mediated endothelial glycolysis promotes pulmonary 144. Xia B, Zhao D, Wang G, Zhang M, Lv J, Tomoiaga AS, Li Y, Wang X, Meng S,
hypertension. Proc Natl Acad Sci U S A. 2019;116:13394–13403. doi: Cooke JP, et al. Machine learning uncovers cell identity regulator by histone
10.1073/pnas.1821401116 code. Nat Commun. 2020;11:2696. doi: 10.1038/s41467-020-16539-4
130. Wu J, Ke X, Fu W, Gao X, Zhang H, Wang W, Ma N, Zhao M, Hao X, Zhang 145. Wang G, Xia B, Zhou M, Lv J, Zhao D, Li Y, Bu Y, Wang X, Cooke JP, Cao
Z. Inhibition of hypoxia-induced retinal angiogenesis by specnuezhenide, Q, et al. MACMIC reveals a dual role of CTCF in epigenetic regulation of
an effective constituent of ligustrum lucidum Ait., through suppression of cell identity genes. Genomics Proteomics Bioinformatics. 2021;21:S1672-
the HIF-1α/VEGF signaling pathway. Molecules. 2016;21:E1756. doi: 0229(21)00057-7. doi: 10.1016/j.gpb.2020.10.008
10.3390/molecules21121756 146. Clayton ZE, Yuen GS, Sadeghipour S, Hywood JD, Wong JW, Huang NF,
131. Cao R, Jensen LD, Söll I, Hauptmann G, Cao Y. Hypoxia-induced retinal Ng MK, Cooke JP, Patel S. A comparison of the pro-angiogenic potential
angiogenesis in zebrafish as a model to study retinopathy. PLoS One. of human induced pluripotent stem cell derived endothelial cells and in-
2008;3:e2748. doi: 10.1371/journal.pone.0002748 duced endothelial cells in a murine model of peripheral arterial disease. Int
132. Rajagopalan S, Mohler ER III, Lederman RJ, Mendelsohn FO, Saucedo J Cardiol. 2017;234:81–89. doi: 10.1016/j.ijcard.2017.01.125
JF, Goldman CK, Blebea J, Macko J, Kessler PD, Rasmussen HS, et al. 147. Sayed N, Wong WT, Ospino F, Meng S, Lee J, Jha A, Dexheimer P, Aronow
Regional angiogenesis with vascular endothelial growth factor in periph- BJ Cooke JP. Transdifferentiation of human fibroblasts to endothelial cells:
eral arterial disease: a phase II randomized, double-blind, controlled study role of innate immunity. Circulation. 2015;131:300–309.
of adenoviral delivery of vascular endothelial growth factor 121 in patients 148. Meng S, Zhou G, Gu Q, Chanda PK, Ospino F, Cooke JP. Transdifferentiation
with disabling intermittent claudication. Circulation. 2003;108:1933–8. requires iNOS activation: role of RING1A S-nitrosylation. Circ Res.
doi: 10.1161/01.CIR.0000093398.16124.29 2016;119:e129–e138.
133. Domigan CK, Warren CM, Antanesian V, Happel K, Ziyad S, Lee S, Krall A, 149. Chanda PK, Meng S, Lee J, Leung HE, Chen K, Cooke JP. Nuclear
Duan L, Torres-Collado AX, Castellani LW, et al. Autocrine VEGF maintains S-nitrosylation defines an optimal zone for inducing pluripotency.
endothelial survival through regulation of metabolism and autophagy. J Cell Circulation. 2019;140:1081–1099.
Sci. 2015;128:2236–48. doi: 10.1242/jcs.163774 150. Cooke JP. Therapeutic transdifferentiation: a novel approach for vascu-
134. McDonald DM, Baluk P. Significance of blood vessel leakiness in cancer. lar disease. Circ Res. 2013;112:748–50. doi: 10.1161/CIRCRESAHA.
Cancer Res. 2002;62:5381–5. 113.301053
135. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, con- 151. Ubil E, Duan J, Pillai IC, Rosa-Garrido M, Wu Y, Bargiacchi F, Lu Y,
sequences, challenges and opportunities. Cell Mol Life Sci. 2020;77:1745– Stanbouly S, Huang J, Rojas M, et al. Mesenchymal-endothelial transition
1770. doi: 10.1007/s00018-019-03351-7 contributes to cardiac neovascularization. Nature. 2014;514:585–590.
136. Leeper NJ, Hunter AL, Cooke JP. Stem cell therapy for vascular regen- doi: 10.1038/nature13839
eration: adult, embryonic, and induced pluripotent stem cells. Circulation. 152. He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, Pu W, Liu Q, Zhang H, Tian
Downloaded from http://ahajournals.org by on July 26, 2023

2010;122:517–26. doi: 10.1161/CIRCULATIONAHA.109.881441 X, et al. Preexisting endothelial cells mediate cardiac neovascularization after
137. Han JK, Chang SH, Cho HJ, Choi SB, Ahn HS, Lee J, Jeong H, Youn SW, injury. J Clin Invest. 2017;127:2968–2981. doi: 10.1172/JCI93868
Lee HJ, Kwon YW, et al. Direct conversion of adult skin fibroblasts to en- 153. Meng S, Lv J, Chanda PK, Owusu I, Chen K, Cooke JP. Reservoir of fibro-
dothelial cells by defined factors. Circulation. 2014;130:1168–1178. doi: blasts promotes recovery from limb Ischemia. Circulation. 2020;142:1647–
10.1161/CIRCULATIONAHA.113.007727 1662. doi: 10.1161/CIRCULATIONAHA.120.046872
138. Rufaihah AJ, Huang NF, Jamé S, Lee JC, Nguyen HN, Byers B, De A, 154. Lee J, Sayed N, Hunter A, Au KF, Wong WH, Mocarski ES, Pera RR,
Okogbaa J, Rollins M, Reijo-Pera R, et al. Endothelial cells derived from Yakubov E, Cooke JP. Activation of innate immunity is required for efficient
human iPSCS increase capillary density and improve perfusion in a nuclear reprogramming. Cell. 2012;151:547–558.
mouse model of peripheral arterial disease. Arterioscler Thromb Vasc Biol. 155. Lai L, Reineke E, Hamilton DJ, Cooke JP. Glycolytic switch is required for
2011;31:e72–9. doi: 10.1161/ATVBAHA.111.230938 transdifferentiation to endothelial lineage. Circulation. 2019;139:119–
139. Huang NF, Niiyama H, Peter C, De A, Natkunam Y, Fleissner F, Li Z, Rollins 133. doi: 10.1161/CIRCULATIONAHA.118.035741
MD, Wu JC, Gambhir SS, et al. Embryonic stem cell-derived endothelial 156. Klarin D, Lynch J, Aragam K, Chaffin M, Assimes TL, Huang J, Lee
cells engraft into the ischemic hindlimb and restore perfusion. Arterioscler KM, Shao Q, Huffman JE, Natarajan P, et al; VA Million Veteran
Thromb Vasc Biol. 2010;30:984–91. doi: 10.1161/ATVBAHA.110.202796 Program. Genome-wide association study of peripheral artery disease
140. Ginsberg M, James D, Ding BS, Nolan D, Geng F, Butler JM, Schachterle in the Million Veteran Program. Nat Med. 2019;25:1274–1279. doi:
W, Pulijaal VR, Mathew S, Chasen ST, et al. Efficient direct reprogram- 10.1038/s41591-019-0492-5
ming of mature amniotic cells into endothelial cells by ETS factors 157. Yoder MC, Annex BH. Does multicolor lineage tracing of endothelial cells
and TGFβ suppression. Cell. 2012;151:559–575. doi: 10.1016/j. provide a black and white answer on clonal expansion in post-natal angio-
cell.2012.09.032 genesis? Circ Res. 2018;122:643–645.
141. Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee SJ, Oh SY, 158. Cooke JP. Inflammation and its role in regeneration and repair. Circ Res.
Tanaka Y, et al. Direct reprogramming of human dermal fibroblasts into 2019;124:1166–1168.

Circulation Research. 2021;128:1944–1957. DOI: 10.1161/CIRCRESAHA.121.318266 June 11, 2021   1957

You might also like