Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Circulation Research

CARDIOVASCULAR ORGANOIDS/3D MODELS REVIEW SERIES

Blood Vessel Organoids for Development and


Disease
Kirill Salewskij , Josef M. Penninger

ABSTRACT: Despite enormous advances, cardiovascular disorders are still a major threat to global health and are responsible for
one-third of deaths worldwide. Research for new therapeutics and the investigation of their effects on vascular parameters is
often limited by species-specific pathways and a lack of high-throughput methods. The complex 3-dimensional environment
of blood vessels, intricate cellular crosstalks, and organ-specific architectures further complicate the quest for a faithful
human in vitro model. The development of novel organoid models of various tissues such as brain, gut, and kidney signified a
leap for the field of personalized medicine and disease research. By utilizing either embryonic- or patient-derived stem cells,
different developmental and pathological mechanisms can be modeled and investigated in a controlled in vitro environment.
We have recently developed self-organizing human capillary blood vessel organoids that recapitulate key processes of
vasculogenesis, angiogenesis, and diabetic vasculopathy. Since then, this organoid system has been utilized as a model
for other disease processes, refined, and adapted for organ specificity. In this review, we will discuss novel and alternative
approaches to blood vessel engineering and explore the cellular identity of engineered blood vessels in comparison to in vivo
vasculature. Future perspectives and the therapeutic potential of blood vessel organoids will be discussed.

Key Words: blood-brain barrier ◼ blood vessels ◼ endothelial cell ◼ extracellular matrix ◼ organoids ◼ vascular diseases
Downloaded from http://ahajournals.org by on October 15, 2023

T
he human vasculature is a complex system respon- it is important to note that there are heterogeneities
sible for maintaining oxygen and nutrient supply between cells of the same type—on the transcriptional as
to tissues, clearing waste products, or shuttling of well as on the functional level. For example, endothelial
immune cells. It precedes the development of most other cells (ECs) form the innermost layer of all vessels. They
organs as it plays a pivotal role in the proper growth and possess specific characteristics which are a function of
differentiation of essentially all tissues in the developing the tissue context: they control permeability of solutes,
embryo.1–4 New blood vessels form via two distinct pro- sense shear stress, and control vasomotor tone.6,7 And
cesses: (1) vasculogenesis, which describes the differ- just as the functions are diverse, it has been shown that
entiation and assembly of mesodermal precursor cells ECs between different tissues have tremendous differ-
(hemangioblasts) into primitive tubular networks (capil- ences in their transcriptional identity.7,8 Through single-
lary plexus) and (2) angiogenesis, which is the process cell RNA sequencing, individual tissue identities of ECs
of subsequent remodeling and branching of new ves- have been identified, mapped to organs, and can now
sels to form a mature network.5 Broadly speaking, the serve as a reference for in vitro studies aiming to recre-
mature blood vessels can be categorized into capillar- ate native, tissue-specific vasculature.9
ies, the venous system, and the arterial system. Arter- Although the tubing of vessels is assembled from ECs,
ies carry the oxygen-rich, high-pressure blood from the the structural integrity and modification of vessel diam-
heart, through arterioles, to tissues where the nutrient eter is controlled by mural cells, that is, pericytes and
exchange is mediated through capillaries. Deoxygenated vascular smooth muscle cells (VSMCs). During angio-
blood is then transported from the capillaries to venules genesis, endothelial tip cells secrete PDGF-B (plate-
and via veins to the lung to be reoxygenated. Although let-derived growth factor subunit B) which is bound by
the vessels belonging to these categories possess clear PDGFRβ (platelet-derived growth factor receptor beta)
differences in their structure and cellular composition, on the surface of developing mural cells, leading to their

Correspondence to: Josef M. Penninger, MD, Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada. Email
josef.penninger@ubc.ca
For Sources of Funding and Disclosures, see page 508
© 2023 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

498   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
intima. The tunica externa or adventitia forms the outmost

3D MODELS REVIEW SERIES


Nonstandard Abbreviations and Acronyms layer and harbors a plethora of different cell types within
a collagen-rich ECM (extracellular matrix), including fibro-
BBB blood-brain barrier blasts, adipocytes, and multiple resident immune cells.15
bFGF basic fibroblast growth factor Besides serving as an anchor for blood vessels to the tis-
BMP-4 bone morphogenetic protein 4 sue, the adventitia contains lymphatic vessels, perivascu-
BVO blood vessel organoid lar nerves, and microvessels (vasa vasorum) that sustain
CTGF connective tissue growth factor the outer layers as luminal diffusion can be insufficient
ECM extracellular matrix
across thick vessels.16 Several studies have shown that
the adventitia is also home to multiple stem- and progen-
ECs endothelial cells
itor-like cells of unknown origin, expressing markers such
EGF epidermal growth factor
as CD34, PDGFRβ, Sca1 (mouse gene stem cell anti-
eNOS endothelial NO synthase gen-1), and Gli1 (GLI family zinc finger 1).17–19 In mouse
IL interleukin models of arteriosclerosis and atherosclerosis, these cells
LDL low-density lipoprotein reside in the adventitia of lesions,20 can acquire VSMC-
PDGF-B platelet-derived growth factor subunit B 17
and osteoblast-like19 identities, and thus contribute to
PDGFRβ platelet-derived growth factor receptor lesions such as vessel calcification. In disease, adventitial
beta fibroblasts can release inflammatory cytokines in vitro21
SCF stem cell factor and differentiate into myofibroblasts promoting calcifica-
sGC soluble guanylate cyclase tion and stenosis in vivo.22,23 Moreover, the vasa vasorum
TNF tumor necrosis factor has been shown to be involved in aneurysm development
VEGF-A vascular endothelial growth factor A and rupture,24,25 neointima formation,26 plaque vascular-
VEGFR vascular endothelial growth factor ization,27 and a proinflammatory positive feedback loop,28
receptor possibly disturbing the immune privilege of the tunica
VSMC vascular smooth muscle cell media.29 Therefore, besides ECs, special attention must
be also paid to perivascular cell types and structures.
Animal models—especially zebrafish—are commonly
recruitment, maturation, and proliferation.10 The arrange- used for vascular studies, but species-specific pathways
ment of mural cells is dependent on the type of vessel. and compensatory mechanisms complicate attempts at
Downloaded from http://ahajournals.org by on October 15, 2023

Capillaries—the smallest vessels in the body—consist modeling disease. A particular example of this is NOTCH3
of ECs, pericytes, and an enwrapping basement mem- (notch receptor 3), a gene in which missense mutations
brane. Arteries possess several layers of VSMCs which cause Cerebral Autosomal Dominant Arteriopathy with
are responsible for the maintenance of vascular tone Subcortical Infarcts and Leukoencephalopathy, which is an
through their contraction and relaxation. A functional inheritable vasculopathy of brain arteries that causes early
crosstalk between the ECs and mural cells is important strokes and dementia.30 To better understand the function
for proper vascular homeostasis and many vascular dis- of NOTCH3, loss of function studies have been performed
orders are a direct result of mural cell dysfunction. As an in animal models. Loss of function of notch3 in zebrafish
example, studies in rats have shown that upon injury to resulted in reduced viability, abnormal morphology of the
the arterial wall, bFGF (basic fibroblast growth factor) vasculature, and a pathological accumulation of blood in
is released from dead cells and leads to proliferation of the head and fins through compromised vessel integrity.31
VSMCs.11 The increase of VSMCs in turn caused intimal However, 2 studies showed that ablation of Notch3 in mice
thickening and promoted atherosclerotic lesions, explain- had no effect on the viability of the embryos.32,33 Although
ing the development of restenosis after angioplasty. the knockout mice also had a pathological vessel pheno-
During thrombosis, capillaries in the brain constrict and type, it was overall milder, suggesting compensatory mech-
cause ischemia in the associated tissue.12 Even if blood anisms in higher vertebrates. Examples like this invoked a
flow is restored, long-lasting reductions in the cerebral need for a human-based in vitro model of the vasculature
blood flow can be observed.13 Experiments on cerebral to overcome such species-specific differences.
cortical slices revealed that upon ischemia pericytes first In the past decade, many attempts have been made
constrict the capillaries and then die, leading to a long- to achieve this goal (Figure 1). The simplest approach to
lasting decrease of the capillary blood flow and loss of the modeling (part of) the vasculature is to culture human
blood-brain barrier integrity.14 Mural and ECs are not the vascular cells as a monolayer and examine phenotypic
only players in pathology. Walls of larger vessels such as or molecular changes caused by genetic perturbations
veins and arteries are comprised of 3 distinct layers. The or under exposure to compounds of interest. Human
innermost layer (tunica intima) consists of ECs forming umbilical vein ECs are a commonly used primary EC
the tubing and the tunica media contains predominantly line for vascular studies. If cultured on a suitable base-
layers of vascular smooth muscle cells that enwrap the ment membrane matrix, these ECs will self-assemble into

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   499


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/
3D MODELS REVIEW SERIES

Figure 1. General overview of different human-based models for vascular studies.


As the complexity of the model and the faithful representation of the in vivo process increases, the ease of handling and scalability usually
decrease. Endothelial cells can be cultured in 2-dimensional (2D) to form networks.34 By using scaffolds or microfluidic systems, more
complex assemblies are achieved—however, they mostly rely on primary cells.39,40 Self-organizing blood vessel organoids (BVOs) capture both
vasculogenesis and angiogenesis.41,42 Integration into microfluidic systems allows for perfusion of the vessels.43 Implant of the organoids into
immunodeficient mice can be performed to achieve larger vessels and a fully perfused human vascular tree, but throughput is low.42 Illustration
credit: Sceyence Studios.

lumenized tube structures that can be quantified and are In recent years, the field of organoids has vastly
amendable for high-throughput analysis.34 Another inter- expanded its repertoire of target tissues. Initially coined
esting element is the ability of ECs to sense and adapt in the year 1907 by Henry Van Peters Wilson based
to flow by changing their orientation. This striking feature on his observations of sponge cells regenerating into a
can be investigated in vitro by culturing cells in a medium whole organism, followed by the observation that primary
of physiological viscosity and using an orbital shaker to mouse mammary epithelial cells form 3-dimensional
induce multiaxial flow.35,36 In 2015, a protocol was pub- structures on a reconstituted basement membrane by Li
lished for the differentiation of induced pluripotent stem et al in 1987, the term organoid nowadays refers to a self-
cells into ECs and VSMCs through Wnt pathway (Wnt/β- organized collection of cells that developed to structurally
catenin pathway) activation by CHIR99021 and subse- and functionally resemble a specific organ.44–46 In syn-
quent exposure to either VEGF-A (vascular endothelial ergy with the discovery of the four Yamanaka factors to
growth factor A) and forskolin or PDGF-BB and Activin induce stem cells from somatic cells, organoids can now
Downloaded from http://ahajournals.org by on October 15, 2023

A.37 Comparison of the gene expression profile of the be used to model genetic disorders through the utiliza-
mural cells demonstrated a high similarity to the respective tion of patient cells.47 In the context of vasculature, there
primary cell type. Indeed, induced ECs achieved a similar are two main themes for the utilization of sophisticated
transendothelial electrical resistance to human umbili- 3-dimensional models. (1) For development and disease,
cal vein ECs and could be subjected to the tube forma- the ability to model and observe interaction of human
tion assay whereas VSMCs responded to stimulation by vascular cells with their environment is indispensable.
vasoconstrictive drugs. Similarly, another protocol reported During angiogenesis, the extracellular matrix is degraded
the derivation of pericytes by magnetically enriching for to allow for migration and infiltration of ECs into the sur-
vascular-induced CD34- cells.38 Culture of these cells in rounding tissue. Stimulation of ECs with proangiogenic
medium containing (amongst others cytokines) EGF (epi- factors leads to upregulation of matrix metalloproteinases
dermal growth factor), bFGF, and VEGF165 yielded a 95% responsible for the degradation of collagens, gelatin, and
pure CD13+/PDGFRβ+ pericyte population that could be fibronectin.48,49 Furthermore, the vasculature is shaped
expanded for at least 10 passages. These differentiation by, and responds to, different physiological and environ-
approaches are very useful for the study of individual vas- mental parameters. Organoid models can therefore (par-
cular cell types and compared to primary lines have the tially) substitute for studies that are either unethical or
advantage that they can be based on patient-derived cells, too resource-intensive to perform on live animals, such
and thus faithfully represent complex disease genotypes. as the effects of hypoxia or modification of flow param-
However, they lack the 3-dimensional environment and eters. (2) A major shortcoming of most organoid culture
tissue assembly observed in vivo. Therefore, specifica- protocols irrespective of the tissue that they represent
tion and invasion of endothelial tip cells into extracellular are the static culture conditions. As organoids surpass a
matrix, codifferentiation and recruitment of mural cells, or certain size, extensive cell death occurs in the core due
vessel response to different flow parameters is difficult to to the physical limits of passive diffusion of nutrients and
study in a 2-dimensional environment. More sophisticated oxygen.50,51 One method of circumventing this limit is by
models that utilize scaffolds or exploit the self-organization culturing organoids in microfluidic chips. The controllable
of vascular cells to form vessels exist, but they cannot fully flow of nutrients not only increases the lifespan of the
capture all developmental stages and thus miss potentially organoids but also leads to an enhancement of organoid
important intermediary cell types39,40 (Figure 1). growth.52,53 Although commercial versions are available,

500   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
most organoid chips are specifically engineered for the we are currently not aware of selective digestion options

3D MODELS REVIEW SERIES


tissue of interest and scalability remains poor. Thus, the for Matrigel. After successful liberation, the organoids
prospect of integrating a functional, perfusable vascula- are usually maintained for approximately 3 to four weeks.
ture into organoids of other tissues has become a key The mature organoids are comprised of ECs, pericytes,
topic. Vascularization of organoids has been shown to mesenchymal stem-like cells, and a very small percent-
contribute positively to maturation of target tissue, and age of hematopoietic cells.42 Thus, for the first time, a
prevascularization of organoids in vitro improved engraft- self-organizing human capillary network was assembled
ment rates upon transplantation in vivo.54–56 For an over- in vitro. These vascular organoids were then used to
view on current strategies to vascularize organoids, we model the process of diabetic vasculopathy. Analysis of
would like to point to the review from Zhang et al.57 Our the dermal vasculature of patients suffering from type
review will focus on current blood vessel organoid (BVO) 2 diabetes showed a thickened and multilayered base-
protocols, their utilization for disease modeling, as well ment membrane. Accordingly, vascular organoids that
as current shortcomings and approaches to solve them. were exposed to elevated levels of glucose, TNF (tumor
necrosis factor), and IL-6 (interleukin 6) in vitro pheno-
copied the expanded basement membrane observed in
BVOS TO STUDY DEVELOPMENT AND
the patient vasculature. The excessive synthesis of ECM
DISEASE was mediated mostly by pericytes, supporting previous
The first protocol for the derivation of a bona fide human observations that mural cells produce and maintain the
BVO was developed by our group in 2019.41,42 This pro- basement membrane.58 Treatment with the γ-secretase
tocol is based on differentiation of multiple vascular cell inhibitor DAPT (N-[N-(3,5-Difluorphenacetyl)-L-alanyl]-
types from a common pool of mesodermal progenitors S-phenylglycin-tert-butylester) abrogated basement
and the intrinsic self-organization of these vascular cells membrane thickening and restored proliferation of ECs.
during development. Stem cells are initially aggregated Finally, pharmacological inhibition and genetic ablation
into embryoid bodies and subsequently pushed into of multiple γ-secretase targets identified DLL4 (delta
mesodermal lineage via Wnt stimulation and BMP-4 like canonical notch ligand 4) and NOTCH3 as the
(bone morphogenetic protein 4). Both mural and ECs key mediators and potential novel targets in diabetic
can be differentiated from a common mesodermal vasculopathy.42
progenitor through supplementation of VEGF-A and In a recent preprint, vascular organoids were
forskolin. After vascular induction, the organoids still used to illustrate the importance of glycolysis in
Downloaded from http://ahajournals.org by on October 15, 2023

resemble a crude aggregate of cells with a diameter ECs and the effects on vasculature upon metabolic
between 100 and 200 microns. To induce sprouting of changes.59 Glycolysis in ECs is mainly regulated by the
vessels, they are embedded into a bilayered ECM sub- enzyme PFKFB3 (6-phosphofructo-2-kinase/fructose-
stitute consisting of a collagen I/Matrigel mix. Of note, 2,6-bisphosphatase 3) and genetic ablation as well
both collagen and Matrigel are sensitive to temperature as pharmacological targeting in mice altered vascular
increases and can polymerize prematurely, leading to sprouting and vessel formation.60 The authors showed
irregular ECM densities and inhibition of sprouting due that upon pharmacological inhibition of PFKFB3 in
to uneven stiffness. Nevertheless, this approach serves organoids, a marked decrease in EC proliferation was
the important purpose of forming a cushion between the observed while pericytes remained unaffected. Further-
organoids and the plastic of the tissue culture ware. The more, the authors reported reductions in vessel den-
density of the organoids is higher than the non-polymer- sity, length, and diameter after 24 hours of treatment,
ized surrounding matrix and without a bilayer assembly, indicating strong vascular remodeling upon glycolysis
they sink upon plating and contact the plastic bottom, inhibition in ECs. Proteomic analysis and confirmation
inhibiting uniform sprouting. via quantitative polymerase chain reaction pinpointed
Upon a few hours of embedding, first cells can be a reduction in CTGF (connective tissue growth factor
seen infiltrating into the ECM. Following this protocol, aka CCN2 [cellular communication network factor 2])
the organoids are cultured for further 4 to 6 days before and subsequent reduced YAP (transcriptional coactiva-
liberation from the Matrigel.41,42 Special attention must be tor YAP1) activity as a possible mechanism. Recombi-
paid to the timing as liberating them too late can cause nant CTGF was able to partially restore YAP activity and
fusions whereas removing them too early yields insuf- rescued the pharmacologically induced vessel pheno-
ficient differentiation indicated by dense cores. The lib- type. The study demonstrated that our vascular organ-
eration from the Matrigel is performed manually under a oids can be indeed used to study metabolic effects on
sterile hood with fine syringe needles. Future studies on vasculature and that perturbation of metabolism has
the optimization of the protocol should focus on alternative marked effects on human blood vessels.
approaches to this step, as it is not only time-intensive but Evidently, there is potential for the vascular organoids
also increases the risk of contamination. Potential solu- to be used for disease research as they show phenotypic
tions could be found through enzymatic treatments, but alterations that are normally restricted to vasculature in

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   501


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/

vivo. Although a great effort has been put into establish- cultures cannot model excessive proliferation of VSMCs,
3D MODELS REVIEW SERIES

ing disease modeling, the developmental progression of infiltration by macrophages, or plaque formation. This
individual cell types and their structural context (venous limitation can be circumvented through transplantation of
versus arterial ECs) remained unknown. In a preprint by the organoids into the renal capsule of immunodeficient
Nikolova et al,61 the authors tackled this problem by per- NOD/SCID (nonobese diabetic/severe combined immu-
forming a comprehensive single-cell transcriptomic analy- nodeficiency) gamma mice. There, the organoids survived
sis of the BVOs at different developmental stages. After for up to 6 months, anastomosed to the host vasculature,
induction of the vascular lineage, PDGFRβ+ pericytes and became fully perfusable as demonstrated through
were detectable already at day 3 of the protocol whereas injection of fluorescent dextran and human-specific anti-
CD31+ ECs arose at day 4, indicating that pericyte devel- CD31 antibodies.42 Through genetic tagging, we further
opment appears to precede maturation of ECs, at least in demonstrated that virtually all pericytes and ECs in these
this organoid model. The transcriptomic identity of ECs in transplants were of human origin. Although many of the
the organoid was dynamic but resembled an in vivo devel- general angiogenic pathways are conserved between
opmental course. A transient upregulation of the EC fate species, there are fine differences in protein isoforms and
transcription factor ETV2 (ETS variant transcription factor expression patterns. As an example, high doses of mouse
2) was seen in a small cell population at day 4, followed VEGF164, but not human VEGF165 are able to induce
by upregulation of common EC markers such as CLDN5 angioma-like structures in SCID mice.69 Importantly, and
(claudin 5) and PECAM1 (platelet and endothelial cell unlike other model systems, it appears that these trans-
adhesion molecule 1). ECs initially adapted a transcrip- planted vessels exclusively utilize human mesodermal
tional identity comparable to arterial ECs until day 14 of cells for the development and recruitment of mural cells
culture, followed by an induction of venous EC markers even to the arteries instead of relying on mouse mural
at day 21. The cause of this identity switch is unknown cells.42 The development into complex vessels could
and should be acknowledged for disease modeling. A enable the study of media- and adventitia-driven disor-
recent study reported a protocol for generation of human ders in a human context. Hopefully, future studies will be
artery- and vein-specific ECs and illustrated the conflicting able to present methods to develop larger vessels without
specification pathways.62 However, it also highlighted the the need for an animal host, thus greatly boosting high-
importance of the 2 identities in disease as two viruses throughput research for arterial- or vein-specific diseases.
preferentially targeted arterial ECs. In our organoids, the In vitro, perfusion of BVOs remains a challenge. The
identity switch could be related to the observed upregula- fact that this (and other organoids) require a trans-
Downloaded from http://ahajournals.org by on October 15, 2023

tion of SULF1 (sulfatase 1) in mural cells.61 SULF1 inhibits plantation into an animal host for proper vasculariza-
EC angiogenesis through attenuation of VEGF-mediated tion and perfusion increases both cost and time while
signaling, which in turn is required for arterial specifica- simultaneously reintroducing potential species-specific
tion of ECs.63–65 Analysis of differentiation trajectories fur- effects. ECs sense changes in flow-induced shear stress
ther revealed that ECs arose from 2 different precursor through a plethora of mechanosensing membrane pro-
pools: although the majority developed from lateral plate teins including integrins, G-protein–coupled receptors,
mesoderm progenitor cells as expected, late organoid and junction proteins.70–72 One of the downstream media-
stages revealed a trajectory from mural cells into ECs.61 tors is the eNOS (endothelial NO synthase) which pro-
Pericytes are known to display a remarkable multipotency duces NO. NO diffuses to VSMCs and enables vascular
through transdifferentiation into other mesenchymal or relaxation through activation of sGC (soluble guanylate
immune cells.66–68 This transdifferentiation could serve as cyclase) and subsequent cGMP production.73 Inadequate
a compensatory mechanism to increase the number of sensing and adaption to shear stress and in particular the
ECs. More studies into pericyte-to-endothelial transdiffer- NO signaling pathway have long been associated with a
entiation are required and could reveal novel mechanistic cohort of vascular disorders.74,75 For instance, mutations
insights in various vascular disorders where abnormal cell in sGC are associated with Moyamoya disease—a steno-
proliferation and differences in pericyte versus EC num- occlusive vascular brain disorder of unknown cause.76
bers are a key element to pathogenesis. Besides shear stress, pressure, and associated strain,
One shortcoming of our capillary organoid protocol which constitute changes in dimension or deformation,
is the inability to generate larger vessels in vitro.42 Inde- act upon the vessel walls.77 Branching points and bifur-
pendent of culture stage, in vitro the vessels are unable cations are often subjected to haphazard hemodynamic
to increase complexity and form arteriole- or venule-like forces, thereby disturbing laminar flow.78 Atherosclerotic
structures. Thus, the organoids also do not possess a lesions predominantly occur at these bifurcation points.79
tunica media or adventitia, confounding in vitro studies Measurements in bovine vessels have demonstrated that
of atherosclerotic processes in these layers. Although branch regions experience double the amount of strain
it should be feasible to study certain disease hallmarks compared to neighboring areas.80 Modulations of blood
such as changes in EC gene expression, altered pericyte pressure through factors such as dietary habits, smok-
coverage, and angiogenic sprouting, current organoid ing, or genetic predisposition can increase the already

502   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
elevated stress in these areas, leading to cumulative only few mural cells could be observed in vitro. Never-

3D MODELS REVIEW SERIES


tissue injury over time.78 Finally, the increased injury theless, the study demonstrated that 3-dimensional vas-
and stretch could lead to augmented LDL (low-density cular structures in an organoid can be achieved without
lipoprotein) penetration,78 supported by in vivo observa- the utilization of a highly variable matrix and prolonged
tions of LDL accumulation predominantly at branching subjection to proangiogenic factors, exploiting the cells’
regions.81 Pressure effects, especially at vascular bifur- intrinsic ability to self-organize given the correct tissue
cation points, are impossible to study in a 2-dimensional context. Future advances should find a compromise
system. To tackle the lack of controllable perfusion, we between both approaches, for example, through fully
are currently working on a microfluidic approach for vas- synthetic hydrogels with specific stiffnesses closely
cular organoids.43 An analysis of flow and shear rates mimicking the organotypic environment of interest.
revealed values comparable to those observed in human
capillaries, thus allowing us to further refine the vascular ORGANOTYPIC VASCULATURE AND
organoid model.43 As a next step, it would be interesting
to study how altered pressure influences vessel remodel- SPECIFIC FUNCTIONS
ing, EC characteristics, or mural cell composition. Besides transcriptional differences, the vasculature in
The importance of flow in vascular diseases has different organs is often highly adapted to execute spe-
been recently also addressed using induced pluripo- cific functions and cellular organizations. One example
tent stem cell-derived ECs from a patient carrying a for this is the arrangement of ECs and EC permeabil-
mutation in ENDOGLIN—resulting in hereditary hem- ity in different organs. Brain ECs are one example for
orrhagic telangiectasia.82 Hereditary hemorrhagic tel- restricted permeability as their tight junctions contribute
angiectasia–EC networks were indistinguishable from to the blood-brain barrier (BBB). In the kidney, a fenes-
control ECs in 2-dimensional culture but upon imple- trated endothelium can be found that allows for passage
mentation into a 3-dimensional and flow environment of larger molecules and is important for glomerular filtra-
on a chip showed reduced vascular densities, EC pro- tion.86 Discontinuous endothelial layers associated with
liferation, and pericyte coverage. a porous basement membrane are found in the liver,
The current BVO protocols require the utilization of spleen, and bone marrow.7 Specialized cell types can
Matrigel as an ECM for vessel sprouting. Matrigel is fre- also associate with the blood vessels and modulate their
quently used as a matrix for culture of many cell types function, such as astrocytes in the brain or hematopoietic
as it is rich in ECM proteins, such as collagen IV, lam- stem cells in the bone marrow. In the following section,
Downloaded from http://ahajournals.org by on October 15, 2023

inin, and entactin, as well as multiple growth factors.83,84 we will discuss efforts to develop organotypic vascular
The exact composition, however, varies from batch to organoids and compare their features to in vivo counter-
batch, and although growth-factor–reduced versions parts (Figure 2 and Table 1).
are manufactured, they still suffer from variability.83 To
address this problem, Schmidt et al85 newly presented
a BVO protocol that abstains from the use of Matrigel. Brain Vasculature
Instead, the authors utilized a conical agarose coating in The brain vasculature is highly specialized: astrocytes
96-well plates for the aggregation of induced pluripo- associate tightly with blood vessels and in conjunction
tent stem cells and subsequent organoid culture. Similar with tight endothelial junctions form the blood-brain bar-
to our initial protocol, a treatment with CHIR99021 and rier. Microglia serve as specialized immune cells, con-
BMP4 was performed for 3 days to induce mesoderm. tribute to neurogenesis and modulate recovery under
However, vascular induction was done through a single injury.91,92 Accordingly, these brain-specific cells can con-
dose of 100 ng/mL VEGF for 48 hours, after which tribute to cerebral vasculopathies; for example, astrocytes
organoids were permanently maintained in N2B27- play a role in the development of cerebral cavernous mal-
medium without further vascular-specific growth factors. formations.93 Mechanistically, upon CCM3 mutations, an
This alternate culture protocol and lack of embedding in endothelial-mediated elevation of NO stabilized HIF-1α
an ECM substitute directly affected the morphology of (hypoxia-inducible factor 1-alpha) in astrocytes and lead
the organoids: while early-stage organoids consisted of to elevated VEGF production and initiation of a quasi-
loosely connected mesenchymal cells, at day 7 a vas- hypoxic program, prompting abnormal angiogenesis.
culogenic zone could be observed.85 Cells in this zone In 2013, Lancaster et al50 published the first protocol
stained positive for CD31 and remodeled after several for brain organoids, and significant advances in struc-
days by infiltrating into the other tissue layers of the ture and complexity have been made since. One criti-
organoid. The identity of the outer tissue is unknown cal aspect for brain organoids is focused on introducing
but has been shown to be GATA6 negative—as opposed vasculature to model the BBB and allow for better nutri-
to the center of the organoid. Although transplantation ent support and hence growth and maturation of larger
into a chick chorioallantois membrane yielded perfusion organoids.94 A study aiming for a 3-dimensional model of
and association of SMA+ mural cells with the network, the BBB exploited the self-organization ability of brain

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   503


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/
3D MODELS REVIEW SERIES
Downloaded from http://ahajournals.org by on October 15, 2023

Figure 2. Illustration of tissues of origin for organotypic blood vessels organoids.


Boxes give information about processes of interest that are modeled by the groups that achieved the organoids. So far, brain-specific, muscle-
specific, and bone marrow–specific vascular organoids have been achieved. Illustration credit: Sceyence Studios.

vascular cells, coseeding primary brain ECs, primary to normal mono- and transwell-cultures, illustrating the
pericytes, and primary astrocytes in hanging drop culture importance of the 3-dimensional environment and the
plates.95 All 3 cell types spontaneously self-assembled ability of pericytes to enhance stable EC interactions.
into spheroids with a distinct cellular organization: ECs Yet, the authors were not able to demonstrate functional
formed the surface layer, astrocytes populated the core, characteristics of a BBB such as selective compound
and pericytes formed an intermediate layer, separating uptake or the presence of tight junctions.
the ECs and astrocytes. Comparing these spheroids A similar protocol to model the BBB in vitro was
to different permutations of cell mixtures, the authors employed with one small but substantial change88:
noticed that ECs are an important component in the reg- VEGF-A was omitted in the spheroid culture medium
ulation of proliferation of the other cell types. The triple- as VEGF-A is known to increase vascular permeabil-
cell spheroids arrested their cell growth after a certain ity by disrupting tight junctions.96,97 This small modi-
time whereas pericyte-astrocyte spheroids grew unhin- fication enabled the spheroid to acquire several BBB
dered until the formation of a necrotic core. Through flow characteristics, illustrating the positive and negative
cytometry analysis, the authors demonstrated a marked effects that a singular growth factor can have on the
increase in surface cell adhesion molecules compared tissue of interest. Following VEGF-A withdrawal, these

504   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
Table 1. Summary of Reported Blood Vessel Organoid Protocols

3D MODELS REVIEW SERIES


Type of ECM for Final medium and growth
Publication vasculature embedding General protocol factors Aim/disease modeling
Wimmer et al Nonorganotypic 75% Collagen I Embedding of mesodermal StemPro-34 SFM Diabetic vasculopathy
201941,42 capillaries solution spheroids in ECM to achieve 100 ng/mL VEGF-A
25% Matrigel sprouting and vessels
100 ng/mL FGF2
15% FCS
Schmidt et al85 Nonorganotypic None Derivation and culture of Single dose of 100 ng/mL Exploitation of intrinsic
capillaries vascular spheroids in conical VEGF-A for 48 h self-organization of vascular
agarose-coated wells N2B27 cells

Romeo et al59 Nonorganotypic 75% Collagen I Embedding of mesodermal StemPro-34 SFM Changes in vascular
capillaries solution spheroids in ECM to achieve 100 ng/mL VEGF-A structure and remodeling
sprouting and vessels upon glycolysis inhibition
25% Matrigel 100 ng/mL FGF2
15% FCS
Nikolova et al61 Nonorganotypic 75% Collagen I Embedding of microwell- StemPro-34 SFM Total characterization of
capillaries solution derived mesodermal spheroids 100 ng/mL VEGF-A development and cellular
in ECM to achieve sprouting identities in BVOs
25% Matrigel and vessels 100 ng/mL FGF2
15% FCS
Dailamy et al87 Muscle- and 2 mg/mL Collagen Individual encapsulation of StemPro-34 SFM Introduction of parenchymal
neural-tissue vascular spheroids in ECM 100 ng/mL VEGF-A cell types into vascular
capillaries organoids through genetic
20% Matrigel 100 ng/mL FGF2 drivers
15% FCS
(20 ng/mL BDNF + 20 ng/mL NT3)
Ulrich et al Brain capillaries 4% Hanging drop spheroid culture EBM -2 Development of
201395 Methylcellulose of primary brain ECs, pericytes, 50 ng/mL VEGF-A 3-dimensional in vitro model
supplementation and astrocytes for the blood-brain barrier
2% FCS
Cho et al88 Brain capillaries None Aggregation of ECs, pericytes, EGM-2/ Endothelial basal Refinement of previous
and primary astrocytes in medium 3-dimensional in vitro model
agarose-coated wells 2% FCS for the blood-brain barrier
Downloaded from http://ahajournals.org by on October 15, 2023

Sun et al54 Brain capillaries Matrigel droplet Embedding of vascular-induced N2B27 Obtaining vascularized
spheroids in Matrigel droplets/ 20 ng/mL VEGF-A brain organoids
brain organoid coculture
Giger et al89 Bone marrow None Microwell-based coculture DMEM/F12 + human endothelial Development of
vasculature of ECs and MSCs to form SFM 3-dimensional bone marrow
spheroids 1x N2 model system/establishing
a model to study leukemia
1x B27 treatments
FGF-2
20 ng/mL EGF
20 ng/mL PDGF-AB
20 ng/mL hOSM
40 ng/mL hIGF-1
15% chick embryo extract
Khan et al90 Bone marrow 30% Collagen I Simultaneous induction of STEMdiff APEL2 Medium Development of
vasculature vascular and hematopoietic 25 ng/mL VEGF-A 3-dimensional bone marrow
commitment with subsequent model system/ TGFβ-
embedding in ECM 25 ng/mL VEGF-C induced bone marrow
25 ng/mL FGF-2 fibrosis of hematological
30% Collagen IV 25 ng/mL BMP-4 cancer

25 ng/mL hSCF
25 ng/mL Flt3
25 ng/mL EPO
40% Matrigel 25 ng/mL TPO
25 ng/mL G-CSF
10 ng/mL IL-3
10 ng/mL IL-6

Organoids are grouped based on the type of vasculature that they represent. A brief description of the utilized ECM substitutes (if any) is included, as well as a description
of the protocol, the final maturation medium, and essential growth factors. The general aim of the study and the modeled diseases are listed. APEL indicates albumin
polyvinylalcohol essential lipids; BDNF, brain-derived neurotrophic factor; BMP, bone morphogenic protein; BVO, blood vessel organoid; EBM, endothelial basal medium; EGM,
endothelial growth medium; EC, endothelial cell; ECM, extracellular matrix; EGF, epidermal growth factor; EPO, erythropoeitin; FCS, fetal calf serum; FGF, fibroblast growth
factor; Flt, fms-like tyrosine kinase; G-CSF, granuolocytic colony-stimulating factor; hIGF, human insulin-like growth factor 1; hOSM, human oncostatin M; IL, interleukin; MSC,
mesenchymal stem cell; NT, neurotrophin; PDGF, platelet-derived growth factor; SFM, serum free media; TPO, thrombopoeitin; VEGF, vascular endothelial growth factor

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   505


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/

spheroids showed the formation of dense tight junc- a discontinuous endothelium that allows passage of cells
3D MODELS REVIEW SERIES

tions between ECs, expression of Claudin and Occlu- and proteins.99,100 The fine balance between differentiating
din and high molecular weight dextran was not able to into the different hematopoietic lineages and self-renewal
diffuse inside the spheroid. The BBB-associated efflux for maintenance of the stem cell pool is critical for many dis-
pump P-gp (ATP-dependent translocase ABCB1 [also eases, such as acute myeloid leukemia. In essence, the bone
known as P Glycoprotein]) and the transcytosis recep- marrow niche represents a special type of blood vessel, and
tor LRP-1 (prolow-density lipoprotein receptor-related recent studies aimed at recapitulating the development of
protein) were also expressed and functional. Expres- this environment in vitro. For instance, Giger et al89 used a
sion levels of P-gp, ZO-1 (tight junction protein ZO-1), microwell-based approach where they coseeded ECs and
and β-Catenin were significantly higher in the spheroid mesenchymal stem cells to form a spheroid. By performing
compared to a transwell model of the BBB. Importantly, several titration experiments with regard to mesenchymal
the authors demonstrated the selective uptake of drugs stem cells:ECs ratios, the authors showed that the number
using matrix-assisted laser desorption/ionization mass of self-renewing mesenchymal stem cells was higher in the
spectrometry imaging.88 Although this model recapitu- dual-cell type organoids and thus depended on the pres-
lates some aspects of the BBB, the cells assemble into ence of ECs. The ECs self-organized into networks which
distinct compartments, partially separated too far away remained stable for about 1 week before showing signs
from each other for uniform interaction. ECs form a layer of decay. Mesenchymal stem cells from these spheroids
on the surface of the spheroid but do not form vessels. could differentiate into adipocytes, chondrocytes, and pre-
Although BVOs with astrocytes and microglia might osteoblasts. Upon seeding of hematopoietic stem cells on 3
be a great model, this has not yet been achieved because days old organoids, the cells homed and migrated towards
of restrictive culture conditions.98 In a recently published the inside of the organoid, which mimics in vivo data where
study, Sun et al54 instead used a coculture of vascular hematopoietic stem cells injected into an irradiated murine
organoids and brain organoids to achieve BBB-like char- host will home to and reconstitute the bone marrow.101
acteristics. The vascular organoids were obtained by an In an alternative approach, induced pluripotent stem cells
initial differentiation towards mesodermal embryoid bod- are initially aggregated and induced towards mesodermal
ies via Wnt signaling and subsequent vascular induction aggregates,90 modifying our initial BVO protocol.41,42 After
using BMP4, bFGF, and later embedding in Matrigel. induction towards vascular and hematopoietic lineage
ECs sprouted and self-assembled into lumenized struc- through addition of BMP4, FGF2, VEGF-A, SCF (stem cell
Downloaded from http://ahajournals.org by on October 15, 2023

tures. Although immunofluorescent staining confirmed factor), and Flt3 (Fms-like tyrosine kinase 3), the organoids
the presence of pericytes, quantitative polymerase chain were embedded in a hydrogel of collagen/Matrigel mix-
reaction for PDGFRβ illustrated an upregulation at day ture. These organoids showed differentiation into lumen-
12 as opposed to day 3 in our original protocol.54,61 ized blood vessels and association of hematopoietic stem
Upon coembedding of a neuroepithelial embryoid body cells to vessels, analogous to the bone marrow. Single-cell
between 2 vascular progenitor organoids, these fusoids sequencing analysis confirmed the presence of several
were further cultured in the neurovascular maturation hematopoietic and stromal cell subtypes. Importantly, tra-
medium. Prolonged culture led to acquisition of several jectory analysis revealed that the differentiated cells arose
BBB characteristics, such as association of pericytes and from the major routes of myeloid differentiation that are
astrocytes with vessels, expression of tight junction pro- also found in vivo.102 Moreover, the authors showed that this
teins CLDN5 and ZO-1, and the efflux transporter P-gp. organoid system can be used to model pathological bone
Similar to the BBB spheroids,88 fluorescent Angiopep-2 marrow fibrosis that occurs during diseases, such as acute
was taken up by the organoid, whereas the scramble leukemia. It would be interesting to see whether the CD34+
control was not, demonstrating selective transcytosis.54 cells obtained from these organoids would be able to repop-
The fact that these BBB characteristics developed is ulate and rescue the bone marrow niche of an irradiated,
surprising considering that the fusion medium contained immunodeficient mouse. Another approach would be a
VEGF-A, suggesting that cellular crosstalks could over- transplantation of such organoids into for instance MISTRG
ride inhibition by extrinsically provided growth factors, (7 modified genes in the genome of these mice: M-CSFh/h
such as VEGF-A. Clearly, such coculture approaches will IL-3/GM-CSFh/h SIRPah/h TPOh/h RAG2-/- IL2Rg-/-)
play a very important role in the coming years as a major mice, which have been modified with human versions of
organoid vascularization strategy and the initial building genes encoding cytokines for innate immune cell devel-
block to build complex organs in vitro. opment and support development of more immune cells
compared to conventional immunodeficient mice.103 Alter-
natively, these bone marrow organoids could be exposed to
Bone Marrow Vasculature controlled flow in microfluidic devices to test if circulation of
The bone marrow is a major source of hematopoietic stem hematopoietic cells can be achieved. Interactions between
cells in the adult human. Hematopoietic stem cells are circulating blood cells and the vasculature are fundamental
found in the vicinity of sinusoids—large quasicapillaries with components in the pathogenesis of many diseases such as

506   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
platelet activation following atherosclerotic plaque rupture. differentiation 1) + BAF60C to establish myovascular

3D MODELS REVIEW SERIES


Future research should focus on incorporating circulating organoids, respectively. Induction of NEUROD1 lead to
blood cells into in vitro vasculature. Overall, great progress MAP2+ (human gene microtubule associated protein
has been made to establish vascularized bone marrow 2) neurons in day 15 BVOs, but they formed large clus-
niches from human stem cells with enormous potential in ters within the organoid instead of distributing uniformly.
disease modeling, generation of hematopoietic progenitors A similar situation was observable for the myovascular
in vitro, or even controlled genetic repair of mutations that organoids as the MYH+ (myosin heavy chain) cells clus-
cause hematopoietic deficiencies. tered in regions instead of distributing in a salt-and-pepper
pattern. Importantly, the authors showed an upregulation
of the neuronal markers MAP2, VGLUT2 (vesicular glu-
Lymphatic Vessels tamate transporter 2), BRN2 (POU class 3 homeobox 2),
Lymphatic vessels represent a specialized subtype of the and FOXG1 (forkhead box G1) upon prolonged culture
microvasculature. Much effort has been put into under- without loss of CDH5 (cadherin 5) or SMA (actin alpha
standing the basic biology behind lymphatic vessels due to 2) expression. Further characterization of the neurovas-
their critical involvement in immunology, tissue drainage, and cular organoids showed a spontaneous firing of neurons,
disease processes.104 Current in vitro approaches include demonstrating, at least using this readout, functional neu-
cell morphology assessment under differential flow and ronal tissue. Subcutaneous implantation of neurovascu-
ECM substrates,105 assays for barrier function,106,107 trans- lar organoids into immunodeficient mice proved that the
port,108 permeability,109 or studies of immune responses organoids were perfusable. The myovascular organoids
to different antigens.110 The process of lymphangiogen- were subjected to chronic electrical stimulation on a chip
esis is of special interest as lymphatic vessels are criti- upon which the embryonic skeletal muscle myosin 3 was
cally involved in tumor metastasis, and cancer cells were upregulated.87 Thus, expression of lineage-specific driver
shown to secrete prolymphangiogenic factors.111 Although genes can serve as a way to introduce parenchymal tis-
the development of blood vessels depends on VEGF-A/ sue and possibly organospecificity into vascular organoids
VEGFR (VEGF receptor)-1/-2 signaling, lymphangio- instead of purely relying on extrinsic growth factors. A major
genesis is modulated by the expression of VEGFR-3 on challenge of this approach is—as the authors themselves
lymphatic ECs, activated by the ligands VEGF-C/-D.104 pointed out—that not all tissues have known genetic drivers
Current models of lymphangiogenesis rely on the utilization for such lineage specification. Alternative approaches such
of lymphatic ECs, either in a scaffold or in coculture with as knockdowns or timed regulation of certain genes—as
Downloaded from http://ahajournals.org by on October 15, 2023

other cells.112,113 To our knowledge, currently there is no in opposed to constitutive overexpression—might be required
vitro model that faithfully recapitulates both lymphvasculo- to engineer complex vascularized tissues of interest.
genesis and lymphangiogenesis. A major hurdle for such
a model is the origin of lymphatic ECs. During embryonic
development, lymphatic ECs arise from a Prox1+(prospero CONCLUSIONS
homeobox 1)/Lyve1+ EC population of veins.114–116 The Vascular diseases constitute a global health burden
requirement of venous vessels—or at least of a venous EC affecting hundreds of millions of people. Recent break-
identity—raises the question of whether currently available throughs in stem cells and tissue engineering have
BVO protocols could be modified to accomplish in vitro allowed the establishment of ECs, pericytes, and also
lymphvasculogenesis. In our BVO model, one can detect bona fide blood vessel organoids under controlled cul-
venous EC markers upon day 21 of culture but the ves- ture conditions. These human BVOs and engineered
sels are of capillary nature and thus might not possess the blood vessel cell types can be used to study disease
required Prox1+/Lyve1+ population.61 A recent protocol processes, normal development, or screen drugs for
described the derivation of human artery- and vein-specific therapeutic effects—one of the major shortfalls of using
ECs from pluripotent stem cells.62 It would be interesting rodents for drug development. However, to fully explore
to test whether these venous ECs have the ability to yield the potential of these organoids, several issues need to
lymphatic ECs and if this approach could be combined with be addressed: utilization of Matrigel, replacements, and
a 3-dimensional differentiation protocol. standardization for mid to high-throughput screens, con-
trolled perfusion, transcriptionally dynamic identities, or
lack of vessel complexity beyond capillaries. However,
Organotypism Through Genetic Drivers several groups have already started to show that these
One organotypic vessel organoid that we would like to dis- issues can be solved. The ability of vessels to form in
cuss is based on overexpression of specific genes to drive absence of Matrigel and protocols for organotypic vascu-
the development of defined cell types.87 In this study, the latures are discussed in this review. These advances and
authors overexpressed NEUROD1 (neuronal differentia- efforts now need to be translated to human medicine
tion 1) to achieve neurovascular, and MYOD1 (myogenic to test whether such organoids can be used for future

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   507


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/

regenerative medicine, including much-needed vascular- 13. Kunz A, Iadecola C. Cerebral vascular dysregulation in the ischemic brain. Handb
3D MODELS REVIEW SERIES

Clin Neurol. 2009;92:283–305. doi: 10.1016/S0072-9752(08)01914-3


ization of other stem cell–derived tissues. 14. Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA,
O’Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regu-
late cerebral blood flow in health and disease. Nature. 2014;508:55–60.
ARTICLE INFORMATION doi: 10.1038/nature13165
15. Tinajero MG, Gotlieb AI. Recent developments in vascular adventitial patho-
Affiliations biology: the dynamic adventitia as a complex regulator of vascular disease.
Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vi- Am J Pathol. 2020;190:520–534. doi: 10.1016/j.ajpath.2019.10.021
enna (K.S., J.M.P.). Vienna BioCenter PhD Program, Doctoral School of the Uni- 16. Boyle EC, Sedding DG, Haverich A. Targeting vasa vasorum dysfunction
versity of Vienna and Medical University of Vienna, Austria (K.S.). Department of to prevent atherosclerosis. Vascul Pharmacol. 2017;96–98:5–10. doi:
Medical Genetics, Life Sciences Institute, University of British Columbia, Vancou- 10.1016/j.vph.2017.08.003
ver, Canada (J.M.P.). 17. Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, Xu Q. Abun-
dant progenitor cells in the adventitia contribute to atherosclerosis of vein
Acknowledgments grafts in ApoE-deficient mice. J Clin Investig. 2004;113:1258–1265. doi:
The authors thank Astrid Hagelkrüys and Gustav Jonsson for their suggestions 10.1172/jci19628
and for proofreading the review. 18. Passman JN, Dong XR, Wu S-P, Maguire CT, Hogan KA, Bautch VL,
Majesky MW. A sonic hedgehog signaling domain in the arterial adventitia
Sources of Funding supports resident Sca1+ smooth muscle progenitor cells. Proc Natl Acad
J.M. Penninger and K. Salewskij have received funding from the Diamond-Black- Sci U S A. 2008;105:9349–9354. doi: 10.1073/pnas.0711382105
fan-Anemia Fundraising (dbaexperiment.org) and the Innovative Medicines Ini- 19. Kramann R, Goettsch C, Wongboonsin J, Iwata H, Schneider R K, Kuppe C,
tiative 2 Joint Undertaking under grant agreement No 101005026. This Joint Kaesler N, Chang-Panesso M, Machado F G, Gratwohl S, et al. Adventi-
Undertaking receives support from the European Union’s Horizon 2020 research tial MSC-like cells are progenitors of vascular smooth muscle cells
and innovation programme and European Federation of Pharmaceutical Indus- and drive vascular calcification in chronic kidney disease. Cell Stem Cell.
tries and Associations (EFPIA). The authors would also like to thank the Leducq 2016;19:628–642. doi: 10.1016/j.stem.2016.08.001
Foundation for supporting their research with the Transatlantic Network of Excel- 20. Torsney E, Mandal K, Halliday A, Jahangiri M, Xu Q. Characterisation
lence grant “ReVAMP — Recalibrating Mechanotransduction in Vascular Malfor- of progenitor cells in human atherosclerotic vessels. Atherosclerosis.
mations” (2022–2027). 2007;191:259–264. doi: 10.1016/j.atherosclerosis.2006.05.033
21. Dutzmann J, Koch A, Weisheit S, Sonnenschein K, Korte L, Haertlé M,
Disclosures Thum T, Bauersachs J, Sedding DG, Daniel J-M. Sonic hedgehog-depen-
J.M. Penninger declares a conflict of interest as he is named on a patent on blood dent activation of adventitial fibroblasts promotes neointima formation. Car-
vessel organoids. Furthermore, he is founder, shareholder, and chairman of the sci- diovasc Res. 2017;113:1653–1663. doi: 10.1093/cvr/cvx158
entific advisory board of Angios Biotech. Angios Biotech is specialized in vascular 22. Li N, Cheng W, Huang T, Yuan J, Wang X, Song M. Vascular adventitia calci-
research based on blood vessel organoids. The other authors report no conflicts. fication and its underlying mechanism. PLoS One. 2015;10:e0132506. doi:
10.1371/journal.pone.0132506
23. Xu F, Ji J, Li L, Chen R, Hu W. Activation of adventitial fibroblasts con-
tributes to the early development of atherosclerosis: a novel hypoth-
REFERENCES esis that complements the ‘Response-to-Injury Hypothesis’ and the
Downloaded from http://ahajournals.org by on October 15, 2023

1. Gao X, Chen X, Taglienti M, Rumballe B, Little MH, Kreidberg JA. Angioblast- ‘Inflammation Hypothesis’. Med Hypotheses. 2007;69:908–912. doi:
mesenchyme induction of early kidney development is mediated by Wt1 and 10.1016/j.mehy.2007.01.062
Vegfa. Development. 2005;132:5437–5449. doi: 10.1242/dev.02095 24. Tanaka H, Zaima N, Sasaki T, Sano M, Yamamoto N, Saito T, Inuzuka K,
2. Leventhal C, Rafii S, Rafii D, Shahar A, Goldman SA. Endothelial tro- Hayasaka T, Goto-Inoue N, Sugiura Y, et al. Hypoperfusion of the adven-
phic support of neuronal production and recruitment from the adult titial vasa vasorum develops an abdominal aortic aneurysm. PLoS One.
mammalian subependyma. Mol Cell Neurosci. 1999;13:450–464. doi: 2015;10:e0134386. doi: 10.1371/journal.pone.0134386
10.1006/mcne.1999.0762 25. Miyata H, Imai H, Koseki H, Shimizu K, Abekura Y, Oka M, Kawamata T, Mat-
3. Dor Y, Camenisch TD, Itin A, Fishman GI, McDonald JA, Carmeliet P, Keshet E. suda T, Nozaki K, Narumiya S, et al. Vasa vasorum formation is associated
A novel role for VEGF in endocardial cushion formation and its potential with rupture of intracranial aneurysms. J Neurosurg. 2019; Aug 16:1–11.
contribution to congenital heart defects. Development. 2001;128:1531– doi: 10.3171/2019.5.JNS19405
1538. doi: 10.1242/dev.128.9.1531 26. Barger AC, Beeuwkes RI, Lainey LL, Silverman KJ. Hypothesis: vasa vaso-
4. Lammert E, Cleaver O, Melton D. Induction of pancreatic differen- rum and neovascularization of human coronary arteries. N Engl J Med.
tiation by signals from blood vessels. Science. 2001;294:564–567. doi: 2010;310:175–177. doi: 10.1056/NEJM198401193100307
10.1126/science.1064344 27. Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E,
5. Coultas L, Chawengsaksophak K, Rossant J. Endothelial cells and Lo K-M, Gillies S, Javaherian K, Folkman J. Inhibition of plaque neovascu-
VEGF in vascular development. Nature. 2005;438:937–945. doi: larization reduces macrophage accumulation and progression of advanced
10.1038/nature04479 atherosclerosis. Proc Natl Acad Sci U S A. 2003;100:4736–4741. doi:
6. Krüger-Genge A, Blocki A, Franke R-P, Jung F. Vascular endothelial cell biol- 10.1073/pnas.0730843100
ogy: an update. Int J Mol Sci. 2019;20:4411. doi: 10.3390/ijms20184411 28. Sedding DG, Boyle EC, Demandt JAF, Sluimer JC, Dutzmann J, Haverich
7. Aird WC. Phenotypic heterogeneity of the endothelium. Circ Res. A, Bauersachs J. Vasa Vasorum Angiogenesis: Key Player in the Initiation
2007;100:158–173. doi: 10.1161/01.res.0000255691.76142.4a and Progression of Atherosclerosis and Potential Target for the Treatment
8. Aird WC. Endothelial cell heterogeneity. Crit Care Med. 2003;31:S221–S230. of Cardiovascular Disease. Front Immunol. 2018;9:706. doi: 10.3389/
doi: 10.1097/01.ccm.0000057847.32590.c1 fimmu.2018.00706
9. Feng W, Chen L, Nguyen PK, Wu SM, Li G. Single cell analysis of endo- 29. Zorc-Pleskovič R, Pleskovič A, Vraspir-Porenta O, Zorc M, Milutinović A.
thelial cells identified organ-specific molecular signatures and heart- Immune cells and vasa vasorum in the tunica media of atheroscle-
specific cell populations and molecular features. Front Cardiovasc Med. rotic coronary arteries. Bosn J Basic Med Sci. 2018;18:240–245. doi:
2019;6:165. doi: 10.3389/fcvm.2019.00165 10.17305/bjbms.2018.2951
10. Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physi- 30. Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG. CADASIL.
ological, and pathological perspectives, problems, and promises. Dev Cell. Lancet Neurol. 2009;8:643–653. doi: 10.1016/S1474-4422(09)70127-9
2011;21:193–215. doi: 10.1016/j.devcel.2011.07.001 31. Zaucker A, Mercurio S, Sternheim N, Talbot WS, Marlow FL. notch3 is
11. Lindner V, Reidy MA. Proliferation of smooth muscle cells after essential for oligodendrocyte development and vascular integrity in zebraf-
vascular injury is inhibited by an antibody against basic fibroblast ish. Dis Model Mech. 2013;6:1246–1259. doi: 10.1242/dmm.012005
growth factor. Proc Natl Acad Sci USA. 1991;88:3739–3743. doi: 32. Domenga V, Fardoux P, Lacombe P, Monet M, Maciazek J, Krebs LT,
10.1073/pnas.88.9.3739 Klonjkowski B, Berrou E, Mericskay M, Li Z, et al. Notch3 is required for
12. Taylor ZJ, Hui ES, Watson AN, Ni X, Deardorff RL, Jensen JH, Helpern JA, arterial identity and maturation of vascular smooth muscle cells. Genes Dev.
Shih AY. Microvascular basis for growth of small infarcts following occlusion 2004;18:2730–2735. doi: 10.1101/gad.308904
of single penetrating arterioles in mouse cortex. J Cereb Blood Flow Metab. 33. Krebs LT, Xue Y, Norton CR, Sundberg JP, Beatus P, Lendahl U, Joutel A,
2016;36:1357–1373. doi: 10.1177/0271678X15608388 Gridley T. Characterization of Notch3-deficient mice: normal embryonic

508   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768


Salewskij and Penninger Blood Vessel Organoids for Development and Disease

CARDIOVASCULAR ORGANOIDS/
development and absence of genetic interactions with a Notch1 mutation. 55. Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang Y-J, Chapeton K,

3D MODELS REVIEW SERIES


Genesis. 2003;37:139–143. doi: 10.1002/gene.10241 Patterson B, Yuan Y, He C-S, et al. Engineering of human brain organoids
34. Kubota Y, Kleinman HK, Martin GR, Lawley TJ. Role of laminin and base- with a functional vascular-like system. Nat Methods. 2019;16:1169–1175.
ment membrane in the morphological differentiation of human endothelial doi: 10.1038/s41592-019-0586-5
cells into capillary-like structures. J Cell Biol. 1988;107:1589–1598. doi: 56. Pham MT, Pollock KM, Rose MD, Cary WA, Stewart HR, Zhou P, Nolta JA,
10.1083/jcb.107.4.1589 Waldau B. Generation of human vascularized brain organoids. Neuroreport.
35. Warboys CM, Ghim M, Weinberg PD. Understanding mechanobiology in cul- 2018;29:588–593:doi: 10.1097/WNR.0000000000001014
tured endothelium: a review of the orbital shaker method. Atherosclerosis. 57. Zhang S, Wan Z, Kamm RD. Vascularized organoids on a chip: strategies for
2019;285:170–177. doi: 10.1016/j.atherosclerosis.2019.04.210 engineering organoids with functional vasculature. Lab Chip. 2021;21:473–
36. Dardik A, Chen L, Frattini J, Asada H, Aziz F, Kudo FA, Sumpio BE. Differ- 488. doi: 10.1039/d0lc01186j
ential effects of orbital and laminar shear stress on endothelial cells. J Vasc 58. Stratman AN, Pezoa SA, Farrelly OM, Castranova D, Dye LE 3rd, Butler MG,
Surg. 2005;41:869–880. doi: 10.1016/j.jvs.2005.01.020 Sidik H, Talbot WS, Weinstein BM. Interactions between mural cells and
37. Patsch C, Challet-Meylan L, Thoma EC, Urich E, Heckel T, O'Sullivan JF, endothelial cells stabilize the developing zebrafish dorsal aorta. Develop-
Grainger SJ, Kapp FG, Sun L, Christensen K, et al. Generation of vascular ment (Camb). 2017;144:115–127. doi: 10.1242/dev.143131
endothelial and smooth muscle cells from human pluripotent stem cells. Nat 59. Romeo SG, Secco I, Schneider E, Reumiller CM, Santos CX, Pooni A, Yin
Cell Biol. 2015;17:994–1003. doi: 10.1038/ncb3205 X, Theofilatos K, Trevelin SC, Zeng L, et al. Human blood vessel organoids
38. Aisenbrey EA, Torr E, Johnson H, Soref C, Daly W, Murphy WL. A protocol reveal a critical role for CTGF in maintaining microvascular integrity. bioRxiv.
for rapid pericyte differentiation of human induced pluripotent stem cells. 2022;2022.09.01.505804. doi: 10.1101/2022.09.01.505804
STAR Protoc. 2021;2:100261. doi: 10.1016/j.xpro.2020.100261 60. de Bock K, Georgiadou M, Schoors S, Kuchnio A, Wong BW, Cantelmo AR,
39. Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen DT, Cohen DM, Toro E, Quaegebeur A, Ghesquière B, Cauwenberghs S, Eelen G, et al. Role of
Chen AA, Galie PA, Yu X, et al. Rapid casting of patterned vascular net- PFKFB3-driven glycolysis in vessel sprouting. Cell. 2013;154:651–663.
works for perfusable engineered three-dimensional tissues. Nat Mater. doi: 10.1016/j.cell.2013.06.037
2012;11:768–774. doi: 10.1038/nmat3357 61. Nikolova MT, He Z, Wimmer RA, Seimiya M, Nikoloff JM, Penninger JM,
40. Chen MB, Whisler JA, Jeon JS, Kamm RD. Mechanisms of tumor cell Camp JG, Treutlein B. Fate and state transitions during human blood
extravasation in an in vitro microvascular network platform. Integr Biol vessel organoid development. bioRxiv. 2022;2022.03.23.485329. doi:
(Camb). 2013;5:1262–1271. doi: 10.1039/c3ib40149a 10.1101/2022.03.23.485329
41. Wimmer RA, Leopoldi A, Aichinger M, Kerjaschki D, Penninger JM. Gen- 62. Ang LT, Nguyen AT, Liu KJ, Chen A, Xiong X, Curtis M, Martin RM, Raftry BC,
eration of blood vessel organoids from human pluripotent stem cells. Nat Ng CY, Vogel U, et al. Generating human artery and vein cells from pluripotent
Protoc. 2019;14:3082–3100. doi: 10.1038/s41596-019-0213-z stem cells highlights the arterial tropism of Nipah and Hendra viruses. Cell.
42. Wimmer RA, Leopoldi A, Aichinger M, Wick N, Hantusch B, 2022;185:2523–2541.e30. doi: 10.1016/j.cell.2022.05.024
Novatchkova M, Taubenschmid J, Hämmerle M, Esk C, Bagley JA, et 63. Narita K, Staub J, Chien J, Meyer K, Bauer M, Friedl A, Ramakrishnan S,
al. Human blood vessel organoids as a model of diabetic vasculopathy. Shridhar V. HSulf-1 inhibits angiogenesis and tumorigenesis in vivo. Cancer
Nature. 2019;565:505–510. doi: 10.1038/s41586-018-0858-8 Res. 2006;66:6025–6032. doi: 10.1158/0008-5472.can-05-3582
43. Quintard C, et al. An automated microfluidic platform integrating functional 64. Casie Chetty S, Rost MS, Enriquez JR, Schumacher JA, Baltrunaite K,
vascularized organoids-on-chip. bioRxiv. 2021;2021.12.29.474327. doi: Rossi A, Stainier DY, Sumanas S. Vegf signaling promotes vascular endothe-
10.1101/2021.12.29.474327 lial differentiation by modulating etv2 expression. Dev Biol. 2017;424:147–
44. Wilson H. A new method by which sponges may be artificially reared. Sci- 161. doi: 10.1016/j.ydbio.2017.03.005
ence. 1907;25:912–915. doi: 10.1126/science.25.649.912 65. Lanner F, Sohl M, Farnebo F. Functional arterial and venous fate is deter-
Downloaded from http://ahajournals.org by on October 15, 2023

45. Li ML, Aggeler J, Farson DA, Hatier C, Hassell J, Bissell MJ. Influence of mined by graded VEGF signaling and notch status during embryonic stem
a reconstituted basement membrane and its components on casein gene cell differentiation. Arterioscler Thromb Vasc Biol. 2007;27:487–493. doi:
expression and secretion in mouse mammary epithelial cells. Proc Natl Acad 10.1161/01.ATV.0000255990.91805.6d
Sci U S A. 1987;84:136–140. doi: 10.1073/pnas.84.1.136 66. Volz KS, Jacobs AH, Chen HI, Poduri A, McKay AS, Riordan DP,
46. Lancaster MA, Knoblich JA. Organogenesisin a dish: modeling development Kofler N, Kitajewski J, Weissman I, Red-Horse K. Pericytes are pro-
and disease using organoid technologies. Science. 2014;345:1247125. doi: genitors for coronary artery smooth muscle. Elife. 2015;4:e10036. doi:
10.1126/science.1247125 10.7554/eLife.10036
47. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from 67. Dellavalle A, Maroli G, Covarello D, Azzoni E, Innocenzi A, Perani L,
mouse embryonic and adult fibroblast cultures by defined factors. Cell. Antonini S, Sambasivan R, Brunelli S, Tajbakhsh S, et al. Pericytes resident
2006;126:663–676. doi: 10.1016/j.cell.2006.07.024 in postnatal skeletal muscle differentiate into muscle fibres and generate
48. Heo SH, Choi YJ, Ryoo HM, Cho JY. Expression profiling of ETS and MMP satellite cells. Nat Commun. 2011;2:499. doi: 10.1038/ncomms1508
factors in VEGF-activated endothelial cells: role of MMP-10 in VEGF-induced 68. Krautler NJ, Kana V, Kranich J, Tian Y, Perera D, Lemm D, Schwarz P,
angiogenesis. J Cell Physiol. 2010;224:734–742. doi: 10.1002/jcp.22175 Armulik A, Browning JL, Tallquist M, et al. Follicular dendritic cells emerge
49. Quintero-Fabián S, Arreola R, Becerril-Villanueva E, Torres-Romero JC, from ubiquitous perivascular precursors. Cell. 2012;150:194–206. doi:
Arana-Argáez V, Lara-Riegos J, Ramírez-Camacho MA, Alvarez-Sánchez ME. 10.1016/j.cell.2012.05.032.
Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol. 69. Mujagic E, Gianni-Barrera R, Trani M, Patel A, Gürke L, Heberer M, Wolff T,
2019;9:1370. doi: 10.3389/fonc.2019.01370. Banfi A. Induction of aberrant vascular growth, but not of normal angio-
50. Lancaster MA, Renner M, Martin C-A, Wenzel D, Bicknell LS, Hurles ME, genesis, by cell-based expression of different doses of human and mouse
Homfray T, Penninger JM, Jackson AP, Knoblich JA. Cerebral organoids VEGF is species-dependent. Hum Gene Ther Methods. 2013;24:28–37.doi:
model human brain development and microcephaly. Nature. 2013;501:373– 10.1089/hgtb.2012.197
379. doi: 10.1038/nature12517 70. Jalali S, del Pozo MA, Chen K, Miao H, Li Y, Schwartz MA, Shyy JY, Chien S.
51. McMurtrey RJ. Analytic models of oxygen and nutrient diffu- Integrin-mediated mechanotransduction requires its dynamic interaction
sion, metabolism dynamics, and architecture optimization in three- with specific extracellular matrix (ECM) ligands. Proc Natl Acad Sci U S A.
dimensional tissue constructs with applications and insights in 2001;98:1042–1046. doi: 10.1073/pnas.98.3.1042
cerebral organoids. Tissue Eng Part C Methods. 2016;22:221–249. doi: 71. Kuchan MJ, Jo H, Frangos JA. Role of G proteins in shear stress-
10.1089/ten.TEC.2015.0375 mediated nitric oxide production by endothelial cells. Am J Physiol.
52. Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, Mau D, 1994;267:C753–C758. doi: 10.1152/ajpcell.1994.267.3.C753
Valerius MT, Ferrante T, Bonventre JV. Flow-enhanced vascularization and 72. Tzima E, Irani-Tehrani M, Kiosses WB, Dejana E, Schultz DA, Engelhardt B,
maturation of kidney organoids in vitro. Nat Methods. 2019;16:255–262. Cao G, DeLisser H, Schwartz MA. A mechanosensory complex that mediates
doi: 10.1038/s41592-019-0325-y the endothelial cell response to fluid shear stress. Nature. 2005;437:426–
53. Nikolaev M, Mitrofanova O, Broguiere N, Geraldo S, Dutta D, Tabata Y, 431. doi: 10.1038/nature03952
Elci B, Brandenberg N, Kolotuev I, Gjorevski N, et al. Homeostatic mini- 73. Denninger JW, Marletta MA. Guanylate cyclase and the ·NO/cGMP
intestines through scaffold-guided organoid morphogenesis. Nature. signaling pathway. Biochim Biophys Acta. 1999;1411:334–350. doi:
2020;585:574–578. doi: 10.1038/s41586-020-2724-8 10.1016/s0005-2728(99)00024-9
54. Sun XY, Ju X-C, Li Y, Zeng P-M, Wu J, Zhou Y-Y, Shen L-B, Dong J, Chen Y-J, 74. Li H, Förstermann U. Nitric oxide in the pathogenesis of vascular dis-
Luo Z-G. Generation of vascularized brain organoids to study neurovascular ease. J Pathol. 2000 Feb;190:244–54. doi:10.1002/(SICI)1096-9896
interactions. Elife. 2022;11:e76707. doi: 10.7554/eLife.76707 (200002)190:3<244::AID-PATH575>3.0.CO;2-8.

Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768 February 17, 2023   509


Salewskij and Penninger Blood Vessel Organoids for Development and Disease
CARDIOVASCULAR ORGANOIDS/

75. Cannon RO 3rd. Role of nitric oxide in cardiovascular disease: focus on the 95. Urich E, Patsch C, Aigner S, Graf M, Iacone R, Freskgård P-O. Multicellular
3D MODELS REVIEW SERIES

endothelium. Clin Chem. 1998 Aug;44(8 Pt 2):1809–19. Erratum in: Clin self-assembled spheroidal model of the blood brain barrier. Sci Rep.
Chem 1998 Sep;44:2070. doi: 10.1093/clinchem/44.8.1809 2013;3:1500. doi: 10.1038/srep01500
76. Hervé D, Philippi A, Belbouab R, Zerah M, Chabrier S, Collardeau- 96. Proescholdt MA, Heiss JD, Walbridge S, Mühlhauser J, Capogrossi MC,
Frachon S, Bergametti F, Essongue A, Berrou E, Krivosic V, et al. Loss Oldfield EH, Merrill MJ. Vascular endothelial growth factor (VEGF) modu-
of α1β1 soluble guanylate cyclase, the major nitric oxide receptor, leads lates vascular permeability and inflammation in rat brain. J Neuropathol Exp
to moyamoya and achalasia. Am J Hum Genet. 2014;94:385–394. doi: Neurol. 1999;58:613–627. doi: 10.1097/00005072-199906000-00006
10.1016/j.ajhg.2014.01.018 97. Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-
77. Campinho P, Vilfan A, Vermot J. Blood flow forces in shaping the vascular induced vascular permeability. Nature. 2005;437:497–504. doi:
system: a focus on endothelial cell behavior. Front Physiol. 2020;11:552. 10.1038/nature03987
doi: 10.3389/fphys.2020.00552 98. Sasai N, Yakura R, Kamiya D, Nakazawa Y, Sasai Y. Ectodermal factor
78. Thubrikar MJ, Robicsek F. Pressure-induced arterial wall stress restricts mesoderm differentiation by inhibiting p53. Cell. 2008;133:878–
and atherosclerosis. Ann Thorac Surg. 1995;59:1594–1603. doi: 890. doi: 10.1016/j.cell.2008.03.035
10.1016/0003-4975(94)01037-d 99. Travlos GS. Normal structure, function, and histology of the bone marrow.
79. Gimbrone MA, Topper JN, Nagel T, Anderson KR, Garcia-Cardeña G. Toxicol Pathol. 2016;34:548–565. doi: 10.1080/01926230600939856
Endothelial dysfunction, hemodynamic forces, and atherogenesis. Ann 100. Kopp HG, Avecilla ST, Hooper AT, Rafii S. The bone marrow vascular niche:
N Y Acad Sci. 2000;902:230–240. doi: 10.1111/j.1749-6632.2000. home of HSC differentiation and mobilization. Physiology. 2005;20:349–
tb06318.x 356. doi: 10.1152/physiol.00025.2005
80. Thubrikar MJ, Roskelley SK, Eppink RT. Study of stress concentration in the 101. Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H,
walls of the bovine coronary arterial branch. J Biomech. 1990;23:15–26. Many A, Shultz L, et al. Dependence of human stem cell engraftment and
doi: 10.1016/0021-9290(90)90365-a repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–
81. Thubrikar MJ, Keller AC, Holloway PW, Nolan SP. Distribution of low density 848. doi: 10.1126/science.283.5403.845
lipoprotein in the branch and non-branch regions of the aorta. Atherosclero- 102. Rosenbauer F, Tenen DG. Transcription factors in myeloid develop-
sis. 1992;97:1–9. doi: 10.1016/0021-9150(92)90045-i ment: balancing differentiation with transformation. Nat Rev Immunol.
82. Orlova V, Nahon DM, Cochrane A, Cao X, Freund C, van den Hil F, 2007;7:105–117. doi: 10.1038/nri2024.
Westermann CJJ, Snijder RJ, van Amstel JKP, Dijke PT, et al. Vascular defects 103. Rongvaux A, Willinger T, Martinek J, Strowig T, Gearty SV, Teichmann LL,
associated with hereditary hemorrhagic telangiectasia revealed in patient- Saito Y, Marches F, Halene S, Palucka AK, et al. Development and func-
derived isogenic iPSCs in 3D vessels on chip. Stem Cell Rep. 2022;17:1536– tion of human innate immune cells in a humanized mouse model. Nat
1545. doi: 10.1016/j.stemcr.2022.05.022 Biotechnol. 2014;32:364–372. doi: 10.1038/nbt.2858
83. Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex protein mixture 104. Jeltsch M, Tammela T, Alitalo K, Wilting J. Genesis and pathogen-
required for optimal growth of cell culture. Proteomics. 2010;10:1886– esis of lymphatic vessels. Cell Tissue Res. 2003;314:69–84. doi:
1890. doi: 10.1002/pmic.200900758 10.1007/s00441-003-0777-2
84. Vukicevic S, Kleinman HK, Luyten FP, Roberts AB, Roche NS, Reddi AH. 105. Ng CP, Helm CLE, Swartz MA. Interstitial flow differentially stimulates
Identification of multiple active growth factors in basement membrane blood and lymphatic endothelial cell morphogenesis in vitro. Microvasc Res.
matrigel suggests caution in interpretation of cellular activity related 2004;68:258–264. doi: 10.1016/j.mvr.2004.08.002
to extracellular matrix components. Exp Cell Res. 1992;202:1–8. doi: 106. Price GM, Chrobak KM, Tien J. Effect of cyclic AMP on barrier function of
10.1016/0014-4827(92)90397-q human lymphatic microvascular tubes. Microvasc Res. 2008;76:46–51. doi:
85. Schmidt S, Alt Y, Deoghare N, Krüger S, Kern A, Rockel AF, Wagner N, 10.1016/j.mvr.2008.02.003
Downloaded from http://ahajournals.org by on October 15, 2023

Ergün S, Wörsdörfer P. A blood vessel organoid model recapitulating 107. Henderson AR, Ilan IS, Lee E. A bioengineered lymphatic vessel model
aspects of vasculogenesis, angiogenesis and vessel wall maturation. Organ- for studying lymphatic endothelial cell-cell junction and barrier function.
oids. 2022;1:41–53. doi: 10.3390/organoids1010005 Microcirculation. 2021;28:e12730. doi: 10.1111/micc.12730
86. Satchell SC, Braet F. Glomerular endothelial cell fenestrations: an integral 108. Triacca V, Güç E, Kilarski WW, Pisano M, Swartz MA. Transcellular
component of the glomerular filtration barrier. Am J Physiol Renal Physiol. pathways in lymphatic endothelial cells regulate changes in sol-
2009;296:F947. doi: 10.1152/ajprenal.90601.2008 ute transport by fluid stress. Circ Res. 2017;120:1440–1452. doi:
87. Dailamy A, Parekh U, Katrekar D, Kumar A, McDonald D, Moreno A, 10.1161/CIRCRESAHA.116.309828
Bagheri P, Ng TN, Mali P. Programmatic introduction of parenchymal cell 109. Sato M, Sasaki N, Ato M, Hirakawa S, Sato K, Sato K. Microcirculation-
types into blood vessel organoids. Stem Cell Rep. 2021;16:2432–2441. doi: on-a-chip: a microfluidic platform for assaying blood- and lym-
10.1016/j.stemcr.2021.08.014 phatic-vessel permeability. PLoS One. 2015;10:e0137301. doi:
88. Cho CF, Wolfe JM, Fadzen CM, Calligaris D, Hornburg K, Chiocca EA, Agar 10.1371/journal.pone.0137301
NYR, Pentelute BL, Lawler SE. Blood-brain-barrier spheroids as an in vitro 110. Wagar LE, Salahudeen A, Constantz CM, Wendel BS, Lyons MM,
screening platform for brain-penetrating agents. Nat Commun. 2017;8:1– Mallajosyula V, Jatt LP, Adamska JZ, Blum LK, Gupta N, et al. Modeling
14. doi: 10.1038/ncomms15623 human adaptive immune responses with tonsil organoids. Nat Med.
89. Giger S, Hofer M, Miljkovic-Licina M, Hoehnel S, Brandenberg N, Guiet R, 2021;27:125–135. doi: 10.1038/s41591-020-01145-0
Ehrbar M, Kleiner E, Gegenschatz-Schmid K, Matthes T, et al. Microarrayed 111. Gomes FG, Nedel F, Alves AM, Nör JE, Tarquinio SBC. Tumor angiogen-
human bone marrow organoids for modeling blood stem cell dynamics. APL esis and lymphangiogenesis: tumor/endothelial crosstalk and cellular/mi-
Bioeng. 2022;6:036101. doi: 10.1063/5.0092860 croenvironmental signaling mechanisms. Life Sci. 2013;92:101–107. doi:
90. Khan AO, Rodriguez-Romera A, Reyat JS, Olijnik A-A, Colombo M, Wang G, 10.1016/j.lfs.2012.10.008
Wen WX, Sousos N, Murphy LC, Grygielska B, et al. Human bone mar- 112. Osaki T, Serrano JC, Kamm RD. Cooperative effects of vascular angiogen-
row organoids for disease modelling, discovery and validation of therapeutic esis and lymphangiogenesis. Regen Eng Transl Med. 2018;4:120–132. doi:
targets in hematological malignancies. Cancer Discov. 2022 Nov 9:CD-22- 10.1007/s40883-018-0054-2
0199. doi: 10.1158/2159-8290.CD-22-0199 113. Gibot L, Galbraith T, Kloos B, Das S, Lacroix DA, Auger FA, Skobe M.
91. Colonna M, Butovsky O. Microglia function in the central nervous system Cell-based approach for 3D reconstruction of lymphatic capillaries in vi-
during health and neurodegeneration. Annu Rev Immunol. 2017;35:441– tro reveals distinct functions of HGF and VEGF-C in lymphangiogenesis.
468. doi: 10.1146/annurev-immunol-051116-052358 Biomaterials. 2016;78:129–139. doi: 10.1016/j.biomaterials.2015.11.027
92. Loane DJ, Byrnes KR. Role of microglia in neurotrauma. Neurotherapeutics. 114. Oliver G. Lymphatic vasculature development. Nat Rev Immunol.
2010;7:366–377. doi: 10.1016/j.nurt.2010.07.002 2004;4:35–45. doi: 10.1038/nri1258
93. Lopez-Ramirez MA, Lai CC, Soliman SI, Hale P, Pham A, Estrada EJ, 115. Alitalo K, Tammela T, Petrova T. Lymphangiogenesis in development and
McCurdy S, Girard R, Verma R, Moore T, et al. Astrocytes propel neurovas- human disease. Nature. 2005;438:946–953. doi: 10.1038/nature04480
cular dysfunction during cerebral cavernous malformation lesion formation. 116. Doh SJ, Yamakawa M, Santosa SM, Montana M, Guo K, Sauer JR, Curran N,
J Clin Invest. 2021;131:e139570. doi: 10.1172/JCI139570 Han K-Y, Yu C, Ema M, et al. Fluorescent reporter transgenic mice for in
94. Cakir B, Park IH. Getting the right cells. Elife. 2022;11:e80373. doi: vivo live imaging of angiogenesis and lymphangiogenesis. Angiogenesis.
10.7554/eLife.80373 2018;21:677–698. doi: 10.1007/s10456-018-9629-2

510   February 17, 2023 Circulation Research. 2023;132:498–510. DOI: 10.1161/CIRCRESAHA.122.321768

You might also like