11
RESEARCH ARTICLE JunB regulates angiogenesis and neurovascular parallel alignment in mouse embryonic skin Yasuo Yoshitomi 1 , Takayuki Ikeda 1 , Hidehito Saito 1 , Yoshino Yoshitake 1 , Yasuhito Ishigaki 2 , Toshihisa Hatta 3 , Nobuo Kato 4 and Hideto Yonekura 1, * ABSTRACT Blood vessels and nerve fibers are often closely arranged in parallel throughout the body. Therefore, neurovascular interactions have been suggested to be important for the development of vascular networks. However, the molecular mechanisms and genes regulating this process remain unclear. In the present study, we investigated the genes that are activated in endothelial cells (ECs) following interactions with neurons during vascular development. Microarray analyses of human primary microvascular ECs co-cultured with mouse primary dorsal root ganglion cells showed that JunB is strongly upregulated in ECs by neurovascular interactions. Furthermore, the forced expression of JunB in ECs stimulated a tip-like cell formation and angiogenesis in vitro and induced vascular endothelial growth factor A (VEGFA) and the pro-angiogenic integrin subunit ITGB3 expression. Moreover, in vivo knockdown of JunB in ECs from developing mouse limb skin considerably decreased the parallel alignments of blood vessels and nerve fibers. Taken together, the present data demonstrates for the first time that JunB plays an important role in the formation of embryonic vascular networks. These results contribute to the molecular understanding of neurovascular interactions during embryonic vascular development. KEY WORDS: JunB, AP-1 transcription factor, Neurovascular interaction, Vascular remodeling INTRODUCTION Blood vessels form vast and highly structured networks that pervade all organs in vertebrates and deliver oxygen, nutrition, biologically active substances and immune cells. These highly structured vascular networks are established by endothelial cells (ECs) and the cellmatrix and cellcell interactions between various cell types, including pericytes, smooth muscle cells and neurons. The ensuing cellmatrix and cellcell interactions are believed to function as guidance cues for EC differentiation and vascular development for establishing vascular networks. The loss of these guidance cues reportedly leads to abnormal vascularization, which causes several vascular diseases (Dorrell and Friedlander, 2006). Andreas Vesalius (15141564) was the first to describe parallel alignments of blood vessels and nerves throughout the body (reviewed in Carmeliet and Tessier-Lavigne, 2005) and since then, the presence of neurovascular parallelism has been widely recognized. Accordingly, morphological and genetic evidence suggests that interaction between vascular and nervous systems is critical to appropriate patterning and branching of vascular networks (Carmeliet, 2003). In recent studies, numerous molecular cues have been associated with guidance of EC tip cells and axonal growth cones, and these have been categorized as attractive and repulsive cues for both EC tip cells and axonal growth cones (reviewed in Wälchli et al., 2015). Hence, vascular patterning seems to depend on the combination of attractive and repulsive cues that control sites and times of developmental processes. In particular, arterial-specific ephrin-B2 (EFNB2) and venous-specific EphB4 regulate the establishment of the arterialvenous plexus and form distinct boundaries between these vessels through repulsive effects (Klein, 2004; Pasquale, 2005; Poliakov et al., 2004). In contrast, attraction between arteries and veins has been associated with apelin and its receptor APJ (also known as APLNR) signaling (Kidoya et al., 2015). More specifically, apelin is secreted from arterial ECs, stimulates receptor APJ-expressing venous ECs and acts as an attractive cue for venous migration toward arteries. Hence, both attractive and repulsive cues contribute to arterialvenous juxtapositional alignments (Kidoya et al., 2015). Guidance cues from the extracellular matrix (ECM) have also been reported, and behaviors of endothelial tip cells and axonal growth cones are reportedly regulated by direct interactions with the ECM (Gerhardt et al., 2003; Ruhrberg et al., 2002). Moreover, endothelial tip cell selection is reportedly regulated by lamininintegrin signaling, which induces the expression of Dll4 in ECs (Estrach et al., 2011; Stenzel et al., 2011). During neurovascular patterning and parallelism, local signals from peripheral nerves reportedly control arterial differentiation and patterning of vascular networks in embryonic mouse limb skin (Mukouyama et al., 2002). Moreover, peripheral nerve-derived VEGFA promotes arterial blood vessel differentiation via NRP1- mediated positive feedback (Mukouyama et al., 2005), although altered vascular patterning did not affect nerve patterns (Bates et al., 2003). Hence, cues for vascular patterning that are mediated by neurovascular interactions (i.e. interaction between the nervous and vascular system) and may only operate from nerves to ECs. Recently, neurovascular alignment was shown to be dependent on the activity of Cxcl12Cxcr4 chemokine signaling. Specifically, peripheral neuron-derived Cxcl12 stimulates EC migration as an attractive cue for the recruitment and alignment of distant vessels with nerves (Li et al., 2013). Hence, several of the neuron-derived factors that are involved in neurovascular interactions have been described, although the ensuing EC-mediated intracellular signaling pathways that lead to neurovascular parallelism have not yet been fully elucidated. Received 2 August 2016; Accepted 10 January 2017 1 Department of Biochemistry, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan. 2 Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku- gun, Ishikawa 920-0293, Japan. 3 Department of Anatomy, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan. 4 Department of Physiology, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan. *Author for correspondence ([email protected]) H.Y., 0000-0002-0010-1237 916 © 2017. Published by The Company of Biologists Ltd | Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303 Journal of Cell Science

JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

RESEARCH ARTICLE

JunB regulates angiogenesis and neurovascular parallelalignment in mouse embryonic skinYasuo Yoshitomi1, Takayuki Ikeda1, Hidehito Saito1, Yoshino Yoshitake1, Yasuhito Ishigaki2, Toshihisa Hatta3,Nobuo Kato4 and Hideto Yonekura1,*

ABSTRACTBlood vessels and nerve fibers are often closely arranged in parallelthroughout the body. Therefore, neurovascular interactions havebeen suggested to be important for the development of vascularnetworks. However, themolecular mechanisms and genes regulatingthis process remain unclear. In the present study, we investigated thegenes that are activated in endothelial cells (ECs) followinginteractions with neurons during vascular development. Microarrayanalyses of human primary microvascular ECs co-cultured withmouse primary dorsal root ganglion cells showed that JunB is stronglyupregulated in ECs by neurovascular interactions. Furthermore, theforced expression of JunB in ECs stimulated a tip-like cell formationand angiogenesis in vitro and induced vascular endothelial growthfactor A (VEGFA) and the pro-angiogenic integrin subunit ITGB3expression. Moreover, in vivo knockdown of JunB in ECs fromdeveloping mouse limb skin considerably decreased the parallelalignments of blood vessels and nerve fibers. Taken together, thepresent data demonstrates for the first time that JunB plays animportant role in the formation of embryonic vascular networks.These results contribute to the molecular understanding ofneurovascular interactions during embryonic vascular development.

KEY WORDS: JunB, AP-1 transcription factor, Neurovascularinteraction, Vascular remodeling

INTRODUCTIONBlood vessels form vast and highly structured networks that pervadeall organs in vertebrates and deliver oxygen, nutrition, biologicallyactive substances and immune cells. These highly structuredvascular networks are established by endothelial cells (ECs) andthe cell–matrix and cell–cell interactions between various cell types,including pericytes, smooth muscle cells and neurons. The ensuingcell–matrix and cell–cell interactions are believed to function asguidance cues for EC differentiation and vascular development forestablishing vascular networks. The loss of these guidance cuesreportedly leads to abnormal vascularization, which causes severalvascular diseases (Dorrell and Friedlander, 2006).Andreas Vesalius (1514–1564) was the first to describe parallel

alignments of blood vessels and nerves throughout the body

(reviewed in Carmeliet and Tessier-Lavigne, 2005) and since then,the presence of neurovascular parallelism has been widelyrecognized. Accordingly, morphological and genetic evidencesuggests that interaction between vascular and nervous systems iscritical to appropriate patterning and branching of vascularnetworks (Carmeliet, 2003).

In recent studies, numerous molecular cues have been associatedwith guidance of EC tip cells and axonal growth cones, and thesehave been categorized as attractive and repulsive cues for both ECtip cells and axonal growth cones (reviewed in Wälchli et al., 2015).Hence, vascular patterning seems to depend on the combinationof attractive and repulsive cues that control sites and times ofdevelopmental processes. In particular, arterial-specific ephrin-B2(EFNB2) and venous-specific EphB4 regulate the establishment ofthe arterial–venous plexus and form distinct boundaries betweenthese vessels through repulsive effects (Klein, 2004; Pasquale,2005; Poliakov et al., 2004). In contrast, attraction between arteriesand veins has been associated with apelin and its receptor APJ(also known as APLNR) signaling (Kidoya et al., 2015). Morespecifically, apelin is secreted from arterial ECs, stimulates receptorAPJ-expressing venous ECs and acts as an attractive cue for venousmigration toward arteries. Hence, both attractive and repulsive cuescontribute to arterial–venous juxtapositional alignments (Kidoyaet al., 2015).

Guidance cues from the extracellular matrix (ECM) have alsobeen reported, and behaviors of endothelial tip cells and axonalgrowth cones are reportedly regulated by direct interactions with theECM (Gerhardt et al., 2003; Ruhrberg et al., 2002). Moreover,endothelial tip cell selection is reportedly regulated by laminin–integrin signaling, which induces the expression of Dll4 in ECs(Estrach et al., 2011; Stenzel et al., 2011).

During neurovascular patterning and parallelism, local signalsfrom peripheral nerves reportedly control arterial differentiation andpatterning of vascular networks in embryonic mouse limb skin(Mukouyama et al., 2002). Moreover, peripheral nerve-derivedVEGFA promotes arterial blood vessel differentiation via NRP1-mediated positive feedback (Mukouyama et al., 2005), althoughaltered vascular patterning did not affect nerve patterns (Bates et al.,2003). Hence, cues for vascular patterning that are mediated byneurovascular interactions (i.e. interaction between the nervous andvascular system) and may only operate from nerves to ECs.

Recently, neurovascular alignment was shown to be dependenton the activity of Cxcl12–Cxcr4 chemokine signaling. Specifically,peripheral neuron-derived Cxcl12 stimulates EC migration as anattractive cue for the recruitment and alignment of distant vesselswith nerves (Li et al., 2013). Hence, several of the neuron-derivedfactors that are involved in neurovascular interactions have beendescribed, although the ensuing EC-mediated intracellular signalingpathways that lead to neurovascular parallelism have not yet beenfully elucidated.Received 2 August 2016; Accepted 10 January 2017

1Department of Biochemistry, Kanazawa Medical University School of Medicine,1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan. 2MedicalResearch Institute, KanazawaMedical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan. 3Department of Anatomy, Kanazawa MedicalUniversity School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa920-0293, Japan. 4Department of Physiology, KanazawaMedical University Schoolof Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan.

*Author for correspondence ([email protected])

H.Y., 0000-0002-0010-1237

916

© 2017. Published by The Company of Biologists Ltd | Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 2: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

In this study, we investigated EC gene regulatory networks thatare triggered by neurovascular interactions during embryonicvascular network formation, and identified JunB as a novelangiogenic regulator of blood vessel patterning and the alignmentwith nerve fibers during vascular development.

RESULTSNeurovascular interactions in developing embryonic mouselimb skinInitially, we characterized interactions of ECs with nerve fibers atvarious stages of mouse embryonic limb skin development, andexamined vascular and nerve network formation from embryonicday (E)9.5 to E17.5 using whole-mount immunohistochemicalstaining (Fig. 1A). In a previous study of neurovascularremodeling, angiogenesis was observed in the primary capillaryplexus on E9.5 and E13.5, and peripheral nerve migration wasobserved on E15.5–17.5 (Mukouyama et al., 2002). In the presentstudy, nerve fibers innervated tissues at E13.5, when remodelingof the pre-existing primary capillary commenced. Subsequently,neurovascular juxtapositional alignments were established

(Fig. 1A), and following nerve fiber innervation at E13.5, thetotal branch lengths of blood vessels were increased (Fig. 1B) withdecreases in individual branch lengths of blood vessels (Fig. 1C).These data indicate that following nerve fiber innervation,vascular growth and branch compaction progress, and maturevascular networks are formed until E17.5. In association withthese vascular developmental stages, the numbers ofneurovascular cell–cell contacts increased transiently at E15.5,and then decreased by E17.5 (Fig. 1D). Moreover, the numbers ofneurovascular parallel branches increased from E13.5 to E17.5(Fig. 1E), suggesting that neurovascular juxtapositional alignmentis triggered by direct contact of ECs with neurons (Fig. 1A–E).

To investigate the molecular mechanisms underlyingneurovascular juxtapositional alignment, we analyzed generegulation in ECs during neurovascular interactions with co-cultured dorsal root ganglion cells (DRGs). Neurons producedelongated axons in the surrounding spaces, and sufficient numbersof ECs were concomitantly attached to neuronal axons (Fig. 1F).

After microarray analyses of gene expression in the ECs, weselected 69 candidate genes that had a >1.2-fold expression change

Fig. 1. Interactions and juxtapositionalalignment of ECs with peripheral nerve fibersduring the development of mouse skinvasculature. (A) Vasculature and nerve fibers ofwild-type C57BL/6mouse limb skin from embryonicday 9.5 (E9.5), E13.5, E15.5 and E17.5embryos were analyzed by whole-mountimmunohistochemistry with antibodies againstmouse CD31 (red) and neurofilament-M (NF,green), and with the nuclear stain DAPI (blue).Scale bars: 100 µm. Positions of visible cell–cellcontacts between ECs and neurons are indicatedwith arrowheads. Closed juxtapositionalalignments of blood vessels and nerve fibers areshown with dotted lines. Total branch lengths (B),individual branch lengths (C), numbers ofneurovascular contacts (D), and numbers ofparallel branches between blood vessels and nervefibers (E) were quantified as described in theMaterials and Methods. After nerve fiber migrationat E13.5, numbers of neurovascular contactstransiently increased and numbers ofjuxtapositional blood vessels with nerves increasedthroughout development. (F) Co-cultures of humanprimary microvascular endothelial cells (HMVECs;ECs) andmouse dorsal root ganglion cells (DRGs).Scale bars: 100 µm. (G) Gene expression profilingof ECs in co-cultures with mouse DRGs wereanalyzed by performing a microarray experiment.Upregulated and downregulated genes in ECs co-cultured with DRGs were categorized according totheir functions using Gene Ontology analyses.Heat maps show genes upregulated anddownregulated in magenta and cyan, respectively.Boxes indicate the results of two independentexperiments. Asterisks indicate genes withpreviously reported roles in angiogenesis(angiogenesis-related genes). Genes that wereanalyzed and discussed in this study arehighlighted in yellow. Quantitative data in B–E showthe mean with individual data points (threeindependent experiments).

917

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 3: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

in two independent neurovascular co-culture experiments.Candidate genes were then categorized and compared with a listof genes reportedly involved in angiogenesis (Fig. 1G), and 59% ofthese were matched with angiogenesis-related genes (Fig. 1G,indicated with asterisks). In agreement, gene ontology (GO)analyses demonstrated a high representation of angiogenesis andvascular development GO categories among candidate genes(Table S2). Among these, CXCR4 was increased by the presenceof neurovascular interactions (Fig. 1G), and was induced byneurovascular interactions as with a CXCL12 receptor in a previousstudy (Li et al., 2013). Neuron-derived VEGFA reportedlystimulates arterial differentiation of ECs via neurovascularinteraction (Mukouyama et al., 2005). In agreement with this, thearterial differentiation marker EFNB2 was upregulated duringneurovascular interactions in the present study (Fig. 1G), and strongVEGFA induction in ECs was observed (Fig. 1G), potentiallyoffering synergenic contributions to arterial differentiation andangiogenesis. Furthermore, ITGB3, which encodes the β3 subunitof integrin αVβ3 and is known to be involved in angiogenesis(Davis and Senger, 2005; Hayashi et al., 2008), was alsosignificantly upregulated (Fig. 1G).In further analyses of central gene regulators in ECs during

neurovascular juxtapositional alignment, the AP-1 transcriptionfactor JunB was the most strongly and consistently upregulatedtranscription factor (Fig. 2A,B). Consistent with this finding, JunBhas been associated with cell lineage specification anddifferentiation in several cell types, including in hematopoieticstem cells (Santaguida et al., 2009), T cells (Son et al., 2011),

osteoblasts (Guo et al., 2014) and skeletal muscle cells (Raffaelloet al., 2010). Moreover, mice that are homozygous null for JunB diebetween E8.5 and E10 due to impaired placental development, andalthough cell proliferation is normal in JunB-null embryos, retardedembryo growth may reflect failure to establish maternal circulation(Schorpp-Kistner et al., 1999). Thus, in further experiments, weexamined the roles of JunB in vascular network formation andneurovascular parallelism.

JunB expression in ECs during embryonic vasculardevelopment and in vitro angiogenesisHerein, we determined JunB expression in ECs during co-culturewith DRGs and confirmed induction by neurovascular interactionsby performing reverse transcription followed by quantitative real-time PCR (RT-qPCR) (Fig. 2A). In addition, we confirmed theexpression of other AP-1 family members, including JUN, JUND,FOS, FOSL1, FOSL2, ATF2, ATF3 and ATF7, which reportedlyform heterodimers with JunB (van Dam and Castellazzi, 2001). Noupregulation among the AP-1 family, except JunB, was observed inECs co-cultured with DRGs (Fig. 2A). In agreement with theRT-qPCR results, JunB promoter activity was upregulated byneurovascular interactions in luciferase reporter assays, asdetermined using by a NanoLuc luciferase construct containingthe human JunB promoter (Fig. 2B). JunB expression in DRGs wasunchanged by co-culture with ECs (Fig. 2C). Moreover, whole-mount immunohistochemical analyses of mouse embryonic limbskin demonstrated that JunB is expressed in ECs, particularlyin those attached to neurons, in contrast to Fos staining (Fig. 2D).

Fig. 2. JunB is upregulated duringneurovascular interactions in vivo, andduring in vitro 3D-collagen angiogenesis.(A) Expression of JunB and its heterodimercounterparts in ECs co-cultured with DRGs wereexamined using RT-qPCR. The data werenormalized to the expression of Gapdh and arepresented as fold changes compared to separatecontrol cultures. (B) Mouse JunB promoter activitiesin ECs were analyzed in monocultures and in co-cultures with DRGs. (C) JunB expression in DRGsco-cultured with ECs was examined using RT-qPCR. Quantitative data in A–C show themean±s.d. (three independent experiments).(D) Whole-mount immunohistochemistry ofEC-tdTomato mouse embryonic limb skin at E15.5and E17.5 using antibodies against JunB (green),and tubulin βIII (Cyan); JunB expression wasobserved in ECs that were in contact with neurons(arrow heads). Scale bars: 100 µm. (E) JunBinduction was observed in tip cell-like ECs atangiognenic frontiers in in vitro 3D-collagenmatrices.

918

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 4: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

In agreement with these results, in vitro 3D angiogenesis assaysshowed strong JunB induction in tip cell-like ECs at invadingangiogenic frontiers (Fig. 2D).

JunB expression in ECs affects EC shapes and the formationof tip cell-like protrusions, and promotes angiogenesisin vitroTo elucidate JunB functions in ECs, we tested the effects of JunBoverexpression and suppression using shRNA gene silencingmethods. In these experiments, basal JunB expression in ECs wasnot detectable under these conditions (Fig. 3A, left panel).Subsequently, knockdown of JunB using an shJunB vector wasconfirmed in JunB-overexpressing HEK293 cells (Fig. 3A, rightpanel). Overexpression and suppression of JunB strongly affectedEC morphology (Fig. 3B). Specifically, JunB-overexpressing ECsexhibited elongated cell shapes with tip cell-like filopodia andruffling membrane formations, whereas JunB-suppressed ECsshowed polygonal and less-elongated cell shapes (Fig. 3C).Annexin V staining confirmed that JunB knockdown does notinduce apoptosis (Fig. S2). In addition, cell widths weresignificantly decreased following JunB overexpression and wereincreased by JunB suppression (Fig. 3C,D). Accordingly, spheroidoutgrowth assays showed that the formation of tip cell-like ECs wasdependent on JunB expression (Fig. 3E). Mixing spheroidoutgrowth assays using control cells and JunB-overexpressingcells with different fluorescent markers showed that JunB-overexpressing ECs preferentially produce tip-like cells comparedto control cells (Fig. 3F). Moreover, in vitro 3D angiogenesis assaysshowed that overexpression of JunB significantly increased thenumbers of cells that migrated into collagenmatrices compared withthose transduced with the vector control, and JunB knockdownsuppressed EC migration into collagen matrices (Fig. 3G). Takentogether, these data show that JunB controls EC cell elongation andcell motility, and positively regulates angiogenesis in collagenmatrices. In contrast, tube formation on Matrigel surfaces wasindistinguishable between JunB-overexpressing and JunB-suppressed ECs, with no significant differences in tube lengths ornumbers of branches (data not shown), suggesting that JunB-mediated angiogenesis is accompanied by cell migration anddegradation of 3D matrices.

In vivo JunB knockdown in ECs from developing mouse limbskin abrogates neurovascular alignmentTo validate our in vitro data, we examined the effects of in vivo JunBknockdown in ECs from embryonic limb skin vasculature. BecauseJunB knockout (KO) mice die by E9.5 due to placentalmalformation (Schorpp-Kistner et al., 1999), analyses of theroles of JunB in vascular network formation at E9.5–17.5 cannotbe achieved in both conventional and EC-specific conditionalJunB KO mice. Therefore, we developed a novel strategy fordevelopmental stage-, position- and cell-specific JunB knockdownusing a combination of in utero lentivirus injections andCre-mediated cell-specific gene knockdown.These experiments were performed using a shJunB lentivirus

vector (Fig. S4A, upper panel) that expresses shRNA and GFP in allinfected cells, allowing positive identification of lentivirus-infectedcells according to GFP expression. In addition, we administered anEC-specific shJunB lentivirus vector (EC-spe-shJunB; Fig. S4A,lower panel), which contains a loxp-STOP-loxp stuffer sequencebetween the EF1 promoter and the GFP-miR-E-backbone-shRNA-JunB-coding sequence (Fellmann et al., 2013; see Materials andMethods section). This EC-spe-shJunB vector expresses shRNA

and GFP only in Cre-expressing cells, and when injected into VE-cadherin-Cre transgenic mice, the loxp-STOP-loxp stuffer sequenceis removed by Cre-mediated recombination only in cells expressingthe vascular EC marker VE-cadherin.

During nerve and vascular development in embryonic mouselimb skin, the capillary primary plexus is formed at around E9.5,and peripheral nerves (PNs) migrate onto the capillary plexus ataround E13.5. After innervations of PNs onto the capillary plexus,mature vascular networks are formed through neurovascularinteractions until E17.5. Accordingly, we administeredlentiviruses via intra-uterine injections into embryo limbs atE12.5, immediately before the onset of neurovascular interactions,and examined limb skin vasculature after embryo growth untilE17.5 (Fig. S4B). Following injection of shJunB lentiviruses (Fig.S4A, upper panel) into mice, lentivirus-infected and shRNA-expressing cells were identified as GFP positive (Fig. S4C). Insubsequent whole-mount immunohistochemical analyses, mosaicpatterns of shJunB expressing and GFP-positive cells were observedin CD31-positive ECs (CD31 is also known as PECAM1) and inother cell types (Fig. S4D). GFP-negative and CD31-positive ECswere also observed in the fields (Fig. S4E). Thus, these mosaicpatterns of infection were used to analyze EC-specific JunBfunctions in comparison with GFP-negative (non-infected) ECs.

We used EC-tdTomato mice for lentivirus injection. EC-tdTomatomice were generated by crossbreeding of VE-cadherin-Cre mice withB6.Cg-Gt(ROSA)26Sortm9(CAG-tdTomato)Hze/J mice. EC-tdTomatomice expressed tdTomato fluorescent protein specifically in CD31-positive ECs (Fig. S1), allowing identification of vascular ECsaccording to tdTomato expression (Fig. 4A). Subsequently, weanalyzed blood vessel formation and neurovascular alignment of limbskin vasculature in shJunB lentivirus-injected embryos (Fig. 4).Although no significant differences in lengths of non-parallelbranches were observed between GFP-positive and -negative bloodvessels (Fig. 4B), lengths ofGFP-positive (JunB knockdown) parallelbranches were significantly less than those of non-GFP parallelbranches (Fig. 4C). These results indicate that parallel neurovascularalignment was inhibited in shJunB-expressing vasculature. Inagreement, the numbers of parallel GFP-positive JunB knockdownbranches were decreased in these animals (see data points in Fig. 4C).Non-target control shRNA injection did not show any effect onbranch length or the proportion of parallel branches, similar to noinjection (data not shown).

In further studies, we compared the number of parallel branchesto the total number of branches between GFP-positive and -negativevessels, and found that percentage of parallel branches as aproportion the total number of branches was significantly decreasedin GFP-positive (JunB knockdown) vessels (Fig. 4D). In furtheranalyses of neurovascular parallelisms in larger blood vessels(>6.6 µm in diameter), the percentage of parallel branches in GFP-positive (JunB knockdown) vessels were dramatically decreased to20%, whereas those in GFP-negative vessels were 60% of the totalnumber of branches (Fig. 4E). Similar results were obtained in miceinjected with EC-specific shRNA (EC-spe-shJunB; Fig. 4F–H),indicating that downregulation of JunB expression disrupts thebalance of EC cell migration and differentiation that is necessary forthe development of vasculature and the morphogenesis required forneurovascular juxtaposition.

Correlations of ECgene regulationwith JunBoverexpressionand neurovascular interactionsTo investigate pathways downstream of JunB, we performedmicroarray gene expression analyses in JunB-overexpressing ECs,

919

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 5: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

Fig. 3. JunB promotes tip cell-like EC formation and in vitro 3D-collagen angiogenesis. (A) JunB was overexpressed or suppressed in ECs throughlentivirus vectors, and expression was confirmed by western blotting with an anti-JunB antibody; Cont, vector control; JunBOE, JunB overexpression; shCont,non-target shRNA control vector; shJunB, JunB-targeted shRNA. Suppression of JunB by shJunB was confirmed in JunB-overexpressing HEK293 cells(right panel). (B–D) ECs were transduced with lentiviruses expressing human JunB or shJunB and morphological changes were monitored. (B) Phase contrastimages. (C) Longitudinal and lateral lengths of cells were measured and plotted. The bar indicates the regression line as calculated by the software R.(D) Quantification of the lateral lengths of JunB-overexpressing and JunB-suppressed ECs (n=5, three independent experiments). (E) EC spheroid sproutingassays. Overexpression of JunB in ECs stimulated the formation of angiogenic tip cell-like protrusions in 3D-collagen angiogenesis assays, whereas suppressionof JunB expression inhibited the formation of tip cells. Results are mean±s.d. (three independent experiments). (F) EC spheroid sprouting assays of control andJunB-overexpressing ECs. Mixed cultures with equal amounts of mCherry-expressing control ECs (Cont, in red) and GFP-expressing control ECs (Cont, ingreen), or GFP-expressing-JunB-overexpressing ECs (JunBOE, in green) were subjected to the tip-cell assay, and the ratio of the number of tip cells for the greencells to the red cells was quantified (right panel, mean±s.d., *P<0.01mCherry-expressing-ECwith GFP-expressing-EC vsmCherry-expressing-ECwith JunBOE-EC). (G) In vitro 3D angiogenesis assays represented in inverted images of DAPI staining. White lines indicate the surface of collagen matrices and dotted linesindicate average migration distance of ECs; overexpression of JunB significantly stimulated angiogenesis into 3D collagen matrices and suppression of JunBresulted in dramatic inhibition of 3D angiogenesis. (n=5, three independent experiments). For the plots in D and G, the box represents the 25–75th percentiles,and the median is indicated. The whiskers show the maximum and minimum values (three independent experiments). Scale bars: 100 µm (B,E,F).

920

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 6: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

and analyzed the expression of the 69 genes that were identified inneurovascular interaction experiments (Fig. 1G). Among these 69genes, 34 gene expression changes were consistent with thoseaccompanying neurovascular interactions (Fig. 5A), and were likelydownstream of JunB signaling pathways that are triggered by directneurovascular interactions.Among candidate molecules, the integrin subunit β3 (encoded by

ITGB3) was previously shown to promote angiogenesis (Davis andSenger, 2005; Hayashi et al., 2008). In agreement, the presentanalyses showed a significant upregulation of ITGB3 in JunB-overexpressing ECs (Fig. 5B), indicating that ITGB3 expressionis activated by JunB following neurovascular interactions.

Accordingly, we also observed the induction of VEGFA, EFNB2and HES1 in JunB-overexpressing ECs (Fig. 5B). In addition,ITGB3 knockdown in JunB-overexpressing cells significantlyreduced angiogenesis activity in 3D collagen matrices (Fig. 6E,F).The integrin αVβ3-specific inhibitor Cyclo (RGDfC) also causedsignificant suppression of JunB-induced angiogenesis (Fig. 6F).

Involvement of JunB in known neurovascular interactionsignaling pathwaysFinally, we investigated relationships between JunB activation andother known signaling pathways during neurovascular interactions.A previous study showed that Cxcl12 is secreted by peripheral

Fig. 4. JunB knockdown in ECs reducesjuxtapositional alignment of bloodvessels with neurons in vivo. Vascularand neuronal network formations wereanalyzed with or without JunB knockdownusing in utero lentiviral infection. (A) Twotypical immunohistochemistry images ofmouse limb skin at E17.5 following in uterolentivirus injections of shJunB to visualizeblood vessels (red), neurons (cyan), andshRNA-JunB-expressing and GFP-positiveblood vessels (green) are shown.Immunohistochemistry (IHC), skeletonizedimages, neurovascular parallel branches,and merged skeletonized images areshown (left through right). Parallelalignments of blood vessels with nervefibers are blue, and shRNA-JunB positiveblood vessels are orange. (B–E) Lengths ofnon-parallel branches (B), lengths ofparallel branches and nerve fiberalignments (C), the percentage of parallelbranch numbers as a proportion of totalbranch numbers (D), and the percentage ofparallel branch numbers as a proportion oftotal branch numbers among larger vessels(E) were quantified and compared betweenGFP-positive [GFP(+), JunB-knockdown,green dots], and GFP-negative [GFP(−),wild-type, red dots] vessels. Quantificationcriteria are listed in Fig. S2. N.S., notsignificant. (F–H) EC-specific JunBknockdown in limb skin following in uteroinjection of EC-spe-shJunB lentivirus in VE-cadherin-Cre mice. Two typical images ofimmunohistochemistry and skeletonizedimages (F) and quantifications (G,H) areshown. Scale bars: 100 µm. Knockdown ofJunB in ECs decreased juxtapositionalalignment of blood vessels with nerve fibers.Quantitative data in C–E,G and H show themean with individual data points (fourindependent experiments). For the plotsin B, the box represents the 25–75thpercentiles, and the median is indicated.The whiskers show the maximum andminimum values (four independentexperiments).

921

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 7: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

neurons and Schwann cells, and stimulates angiogenesis andvascular remodeling via the Cxcl12 receptor Cxcr4 during theformation of embryonic vascular networks (Li et al., 2013). Thus,we examined the involvement of JunB in Cxcl12-induced 3Dangiogenesis in vitro (Koh et al., 2008) as described in Fig. 3D. Inthese experiments, the presence of Cxcl12 significantly activatedangiogenesis as previously reported (Fig. 6A), but did not induceJunB expression in EC cultures (Fig. 6B), suggesting that JunB isnot regulated downstream of the Cxcl12 pathway.Subsequently, we determined whether neurovascular interactions

in this system were mediated by soluble factors or by direct cell–cellinteractions using analyses of the effects of conditionedmedium fromDRGs on JunB expression inECs.However, JunB expressionwas notaffected by the addition of DRG conditioned medium in RT-qPCR(Fig. 6C) or JunB promoter assays (Fig. 6D). These results indicatethat induction of JunB during neurovascular interactions of ECs isdirectly triggered by cell–cell or cell–ECM interaction.

DISCUSSIONIn the present study, we demonstrated that JunB is induced in ECs byneurovascular interactions, and that it regulates neurovascularjuxtapositional alignments during embryonic vascular development.The presentwhole-mount immunohistochemical analyses showed thatneurovascular interactions started at E13.5, and neurovascularjuxtapositional alignments were established during E13.5–17.5(Fig. 1). Moreover, neurovascular juxtapositional alignments weretriggered by the direct contact of ECs with neurons (Fig. 1A). Thus, tomodel in vivoneurovascular interactions and investigate theunderlyingmolecular mechanisms, we analyzed changes in EC gene expressionduring co-culturewithDRGneurons.The ensuingmicroarrayanalysesidentified 69 genes that were significantly regulated under theseconditions, and JunB was the most differentially expressed

transcription factor, indicating that it had roles in neurovascularinteractions during vascular network formation (Figs 1G and 2A).

Further whole-mount immunohistochemistry analyses of mouseembryonic limb skin confirmed increased expression of JunB in ECsthat were attached to neurons at E13.5 and E17.5 (Fig. 2C), andstrong JunB induction was observed in tip cell-like ECs at invadingangiogenic frontiers in the in vitro 3D angiogenesis assays (Fig. 2D).Additionally, EC morphologies were found to be dependent onJunB expression levels (Fig. 3), indicating that JunB controls ECshape and motility. Accordingly, in vitro 3D angiogenesis wasdependent on JunB expression levels (Fig. 3), further indicating thatJunB positively regulates angiogenesis.

In accordance with the present in vitro data, in vivo JunBknockdown in the ECs of embryonic limb skin vasculature justbefore the onset of neurovascular interactions significantlydecreased the numbers and lengths of parallel branches withneurons (Fig. 4). Moreover, the percentage of parallel branches as aproportion of the total branches was significantly decreased inJunB-knockdown vasculature (Fig. 4), and these effects weregreater in blood vessels of over 6.6 µm in diameter (Fig. 4E). Takentogether, these data demonstrate the necessity of JunB for bloodvessel formation and neurovascular juxtaposition.

Further microarray analyses showed that interactions with DRGsinduced arterial differentiation of ECs, as indicated by upregulationof EFNB2 (Fig. 1G), suggesting that neuron-derived VEGFAinduces arterial differentiation of ECs, as reported previously (Liet al., 2013). In the present study, neurovascular interactionsinduced VEGFA expression in ECs (Fig. 1G), indicating self-activation of arterial differentiation and angiogenesis followingneurovascular interactions. VEGF induction was also observed inJunB-overexpressing ECs (Fig. 5), further implicating JunB in theinduction of VEGFA in ECs during neurovascular interactions.

Fig. 5. Gene expression analysis of JunB-overexpressingECs. (A) Gene expression changes in JunB-overexpressingECs were analyzed by performing microarray experiments. Heatmaps represent upregulated (magenta) and downregulated(cyan) genes in JunB-overexpressing ECs, and are presentedwith the categorized list of genes associated with neurovascularinteractions (see Fig. 1G). Genes that were upregulated anddownregulated by neurovascular interactions (see Fig. 1H) areindicated by ‘up’ and ‘down’, respectively. Genes arecategorized according to their functions. Genes that wereanalyzed and discussed in this study are highlighted in yellow.(B) Expression levels of JUNB, VEGFA, EFNB2, HES1 andITGB3 in JunB-overexpressing (JunBOE) ECs were analyzedusing RT-qPCR. Data show the mean±s.d. (three independentexperiments).

922

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 8: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

Li et al. reported that neurovascular interactions activate VEGFAand Cxcl12 production in neurons, and that neuron-derived VEGFAinduces ECdifferentiation, whereas Cxcl12 induces angiogenesis andvascular remodeling (Li et al., 2013). Similarly, the present data showthat neurovascular interactions activate VEGFA expression in ECs(Fig. 1G), likely contributing synergistically to EC differentiation,vascular remodeling and activation of angiogenesis. In addition,VEGFA was upregulated in JunB-overexpressing ECs (Fig. 5),suggesting that VEGFA is induced by JunB during neurovascularinteractions.

JunB-null mice die prior to E9.5 due to placental dysplasia(Schorpp-Kistner et al., 1999). Hence, JunB KOmay cause vascularmalformation and abnormal placental vasculogenesis, leading toembryonic lethality. Accordingly, embryonic lethality was threetimes more frequent in shJunB lentivirus-injected embryos than incontrol shRNA-injected embryos (data not shown). In addition,limb malformation and shortness was often observed followingshJunB injections, likely reflecting failure of vascular development(data not shown).

Roles for JunB in vascular development have been reportedpreviously, and JunB is known to activate Foxo1 by inducing miR-182, contributing to lymphatic vascular development in zebrafish.In agreement, loss of JunB in zebrafish resulted in the formation ofparachordal lymphangioblasts (Kiesow et al., 2015), and Foxo1expression reportedly effected EC migration, sprouting and tubeformation (Potente et al., 2005). However, the present microarrayanalysis showed no upregulation of Foxo1 during neurovascularinteractions (data not shown), suggesting that the JunB–miR-182–Foxo1 axis is not involved in JunB-mediated neurovascularjuxtapositional alignment.

Gene expression analyses of JunB-overexpressing ECs co-identified about half of those that were regulated by neurovascularinteractions (Fig. 5), indicating considerable involvement of JunBsignaling in neurovascular interactions. Integrins play importantroles in cell adhesion and migration (Desgrosellier and Cheresh,2010), and ECs express several integrin heterodimers, includingαVβ3, α5β1 and αvβ5 (Plow et al., 2000). In particular, integrinαVβ3 is a critical angiogenic regulator for neovascularization thatacts by co-operating with VEGFR2 (Somanath et al., 2009).Integrin αVβ3 is expressed at lower levels in quiescent ECs, but it issignificantly upregulated in angiogenesis (Leu et al., 2002).Functional blocking of αVβ3 results in defects pertaining toneovasculature without affecting the normal pre-existingvasculature (Brooks et al., 1994a,b). Thus, integrin αVβ3 is a keyregulator of angiogenesis. In the present experiment, we observedthe upregulation of ITGB3, which encodes the β3 subunit of integrinαVβ3, during neurovascular interaction (Fig. 1G).We also observedthe upregulation of ITGB3 in JunB-overexpressing ECs (Fig. 5),indicating that the upregulation of ITGB3 during neurovascularinteraction is mediated by JunB. Moreover, ITGB3 knockdown andintegrin αVβ3-specific inhibition significantly suppressed JunB-induced 3D angiogenesis (Fig. 5D), suggesting that integrin αVβ3is one of the downstream effector molecules in JunB signaling. Thenotch signaling protein HES1 reportedly regulates vascularremodeling and arterial specification of ECs during thedevelopment of brain vasculature (Kitagawa et al., 2013). HES1was reportedly regulated by JunB via direct binding to the HES1promoter (Santaguida et al., 2009). These results suggest thatvascular remodeling and formation of neurovascular parallelism aremediated in part by integrin and/or notch signaling pathways.

Neuron-derived Cxcl12 is known to stimulate angiogenesis andvascular remodeling following binding of its receptor Cxcr4 duringthe formation of embryonic vascular networks (Li et al., 2013).However, the present data do not indicate that JunB is regulateddownstream of the Cxcl12 pathway during neurovascularinteractions (Fig. 6A,B), and instead suggest angiogenic roles forJunB that are independent of the Cxcl12 pathway under theseconditions. Cxcl12 was upregulated in ECs upon JunBoverexpression (Fig. 5B), and was downregulated byneurovascular interaction in ECs (Fig. 1G). We need to clarify themechanisms for these differences in gene regulation of Cxcl12between ECs and neurons in future studies.

Fig. 6. JunB functions in known pathways of neurovascular interactioninduced angiogenesis. (A) Effects of Cxcl12 on 3D-collagenangiogenesis (n=5). (B) RT-qPCR expression analysis of JunB followingCxcl12-induced angiogenesis. (C,D) Effects of conditioned medium fromDRG cell cultures on JunB expression (C) and JunB promoter activity (D) inECs; soluble factors in DRG conditioned medium did not activate JunBexpression in ECs. (E,F) Effects of ITGB3 knockdown in JunB-overexpressing ECs and the integrin αVβ3-specific inhibitor Cyclo (RGDfC)were analyzed in 3D-collagen angiogenesis. Quantitative data in B–E showthe mean±s.d. (three independent experiments). For the plots in A and F,the box represents the 25–75th percentiles, and the median is indicated.The whiskers show the maximum and minimum values (three independentexperiments).

923

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 9: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

In summary, we present several lines of evidence for a novelmechanism involving the induction of the transcription factor JunBduring neurovascular interactions, and subsequent regulation ofangiogenesis toward arterial differentiation and neurovascularjuxtapositional alignment. Specifically, neurovascular interactionsinduced JunB expression in ECs of the primary capillary plexus, andthe resulting activation of pro-angiogenic genes, such as VEGFAand ITGB3, led to the morphological changes of ECs, withformation of tip cell-like cells and migration toward nerve fibers,and establishment of mature neurovascular aligned blood vessels.Although the present experiments reveal novel aspects ofneurovascular interactions during the formation of vascularnetworks, further studies are necessary to confirm therelationships between JunB and other signaling pathways knownto be associated with neurovascular interactions, and to define themechanisms of JunB induction. Nonetheless, the present studyprovides clues for future studies regarding the regulatorymechanisms involved in embryonic vascular development.

MATERIALS AND METHODSCell culture and animalsHuman primary microvascular ECs (HMVECs) were isolated from newbornforeskins (Lifeline Cell Technology, Frederick,MD) and were maintained inHuMedia-EB2 medium supplemented with 5% (v/v) fetal bovine serum(FBS), 5 ng/ml basic fibroblast growth factor (bFGF), 10 μg/ml heparin,10 ng/ml epidermal growth factor (EGF), 1 μg/ml hydrocortisone, and39.3 μg/ml dibutyryl cAMP according to the manufacturer’s instructions(Kurabo, Tokyo Japan). Telomerase immortalized HMVECs (TIME cells;#CRL4025, ATCC, Manassas, VA) were maintained in M131 mediumsupplemented with MVGF growth supplements (Invitrogen, Carlsbad, CA).Dorsal root ganglion cells (DRGs) were isolated from C57BL/6 mice onpostnatal day 3 and were dissociated for 10 min at 37°C using 1-mg/mlcollagenase and 5-mg/ml Dispase in HBSS, and were then cultured on poly-D-lysine-coated plates. Cultures were grown in neurobasal mediumsupplemented with B27 and 10-ng/ml NGF (BD Biosciences, FranklinLakes, NJ), 100-ng/ml penicillin, and 100-U/ml streptomycin. Prior to ECand DRG co-culture experiments, DRGs were grown in six-well plates, anddetached HMVECs were inoculated onto DRG cultures in EC medium at aEC:DRG ratio of 10:1. Co-cultures were then incubated for 8 h and RNAwas extracted. This in vitro co-culture may mimic the initial attachment ofECs with neurons, which causes gene expression changes in ECs. Forseparate cultures, ECs and DRGs were cultured separately in EC mediumand incubated for 8 h in parallel with neurovascular co-cultures. After 8 h,RNAwas extracted separately from ECs and DRGs. RNA corresponding tothe number of each cell type in the co-culture was mixed in the same amountby weight, and used as a separate control culture sample in the microarrayanalysis. Conditioned medium from DRGs or NIH-3T3 fibroblasts wasprepared by culturing cells for 24 h in serum-free M131 medium.

An EC-specific tdTomato fluorescent protein-expressing mouse (EC-tdTomato mouse) was generated by crossing VE-cadherin-Cre mice fromthe National Institute of Biomedical Innovation (NIBIO), JAPAN(repository ID: NIBIO132; Kogata et al., 2006) with B6.Cg-Gt(ROSA)26Sortm9(CAG-tdTomato)Hze/J mouse from the Jackson Laboratory, ME(repository ID :007909; Madisen et al., 2010). Subsequently, tdTomatoexpression in ECs was confirmed in CD31-positive ECs by FACS analysiswith APC-labeled anti-mouse-CD31 antibody (MEC13.3, BioLegend, SanDiego, CA; Fig. S1). Animal procedures were performed in accordance withthe Kanazawa Medical University Animal Guidelines under the authority ofproject license #2016-43. Animals were analyzed regardless of gender.

Plasmid construction and lentivirus productionTo prepare lentiviral constructs, the coding region of human JunB (RefSeqNM_002229) was amplified by RT-PCR using total RNA from HMVECsand was cloned into BamHI and NotI sites of the pCDH vector (SystemBiosciences, Palo Alto, CA), and the sequences were then verified. shRNAvectors were constructed in the pSIH vector (System Biosciences) using

synthetic oligonucleotides against human and mouse JunB antisensesequences (shJunB1, 5′-AGGCTCGGTTTCAGGAGTTTGTAGTCGTG-3′ or shJunB2, 5′-TTGAGCGTCTTCACCTTGTCCTCCAGGCG-3′) andhuman ITGB3 antisense sequences (shITGB3-1, 5′-TACTGGAATCTG-ACGACACAGTCATCCTC-3′ or shITGB3-2, 5′-AAGACAGGTCCATC-AAGTAGTAGATGTCC-3′) with loop 5′-TCAAGAG-3′. A syntheticDNA containing the non-target sequence 5′-GCACTACCAGAGCTAAC-TCAGATAGTACT-3′ carrying the same loop was used as the negativecontrol (sh-Vec). Tissue-specific JunB knockdown in mice was achieved byusing the lentivirus (EC-spe-shJunB) containing the modified miR30-(miR-E-) miroRNA backbone shRNA (Fellmann et al., 2013) withloxp-stop-loxp stuffer sequence. In brief, the mouse JunB antisensesequence 5′-TTTCAGGAGTTTGTAGTCGTG-3′ with a 5′-TAGTGAA-GCCACAGATGTA-3′ loop for shRNA-JunB was inserted into the miR-E-microRNA backbone (miR-E-shJunB). miR-E-shJunB was cloned into thepCDH vector downstream of the GFP open reading frame (ORF). The loxp-stop-loxp fragment was then inserted between EF1 promoter and the GFPtranscriptional start site to allow induction of GFP and shRNA expressionupon Cre-mediated recombination in Cre expressing cells. Lentivirusparticles were then produced in HEK293TN cells (System Biosciences)following transfection of the above constructs, the psPAX2 packagingvector and the pMD2.G envelope vector [deposited by Didier Trono,Laboratory of Virology and Genetics, École Polytechnique Fédérale deLausanne, Switzerland; Addgene plasmid #12259 and #12259 respectively,Addgene, MA]. Supernatants from 48 h culture media were then collectedand lentiviral particles were concentrated using PEG-it solution (SystemBiosciences). Lentivirus infectious units (IFU) were determined accordingto the number of GFP-positive ECs as assessed with serial dilutions oflentivirus from cells at 24 h post transduction.

Microarray analysisTotal RNA was extracted using TRIzol reagent (Invitrogen) and purifiedusing RNeasy Mini kits (Qiagen, Valencia, CA). RNA quality wasassessed according to 28S/18S ratios of rRNA bands on electrophoresisgels under denaturing conditions. Subsequently, 100-ng aliquots of totalRNA were labeled according to the manufacturer’s instructions for theGeneChiP WT Sense Target Labeling kit (Affymetrix, Santa Clara, CA).Fragmented and labeled cDNAs were then hybridized onto AffymetrixGenechip Human Gene 1.0 ST or human U133 plus 2.0 arrays. Arrayswere then washed and stained using the GeneChip Fluidics Station 450and scanned using a 3000 7G GeneChip Scanner (Affymetrix). Allarrays passed the quality control criteria of the Expression Consolesoftware (Affymetrix). Raw data CEL files were then normalized usingthe RMA algorithm and the data exported using Expression Console orGene Spring software (Agilent Technologies, Santa Clara, CA). Geneswith a >1.2-fold change in transcription in two independent experimentswere selected, and non-annotated genes and functionally unknown geneswere excluded. Gene ontology (GO) analyses of genes with significanttranscriptional changes during neurovascular interactions wereperformed using the human reference list in the PANTHER v10.0Enrichment analysis program provided by the Gene Ontologyconsortium (http://pantherdb.org/).

RT-qPCRTotal RNAwas extracted from cells as described above. Subsequently, 1-µgaliquots of total RNA samples were reverse transcribed using PrimeScriptRT reagent Kit (TaKaRa, Otsu, Japan). qPCR was performed using theresulting cDNAs with an initial denaturation at 95°C for 30 s, followed by45 cycles of 95°C for 5 s and 60°C for 30 s using the SYBR Green Premix(Takara) with the primers listed in Table S1 in a StepOnePlus Realtime-PCRSystem (Applied Biosystems, CA).

Western blottingCells were suspended in Laemmli-sample buffer containing 5%2-mercaptoethanol and were sonicated for 30 s and heated at 98°C for5 min. The resulting protein samples were subjected to SDS-PAGE using4–12% polyacrylamide gels (Wako, Osaka Japan). After electrophoresis,proteins were transferred onto PVDF membranes (Invitrogen) and blocked

924

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 10: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

with 5% skimmed milk in Tris-buffered saline. Anti-JunB (G53, cat. no.3746, 1:1000; Cell Signaling Technology, Danvers, MA) and anti-β-actin(cat. no. A2228, 1:4000; Sigma-Aldrich) antibodies were used as primaryantibodies, and anti-rabbit IgG-HRP (Amasham GE, MA) was used as asecondary antibody. Protein signals were detected using ECL PlusChemiluminescent Substrate (Thermo Scientific, Rockford, IL).

In vitro 3D collagen matrix angiogenesis assaysAngiogenic activities of ECs were analyzed using three-dimensional (3D)collagenmatrix angiogenesis assays asdescribedpreviously (Kohet al., 2008).Briefly, type I collagen from rat tails (Millipore, Billerica, MA) wasreconstituted in M199 medium supplemented with 1-µM sphingosine 1-phosphate (S1P; Avanti Polar Lipids, AL) with or without 200 ng/mlrecombinant human Cxcl12 (R&D Systems, Minneapolis, MN) and was thenplaced in the bottom chamber of a µ-Slide (ibidi, Martinsried, Germany) andwas transformed into a gel by incubation at 37°C in a CO2 incubator. DetachedECswere thencounted and re-suspended inM199mediumcontaining reducedserum supplement II at a dilution of 1:250, 40 ng/ml FGF-2, 50 µg/ml ascorbicacid and 50 ng/ml 12-O-tetradecanoylphorbol-13-acetate (TPA). To analyzethe involvement of integrin αVβ3 in JunB-induced 3D collagen angiogenesis,Cyclo (RGDfC) peptide (AnaSpec, CA), an inhibitor for integrin αVβ3-mediated cell attachment,was added in collagengelmatrices at a concentrationof 5 µM. Subsequently, 2×104 cells were plated on the upper layer of theµ-Slide and were incubated in a CO2 incubator for 48 h. Cells were fixed andthen stained with anti-human CD31 (WM59, cat no. 303101, 1:200;BioLegend), anti-JunB (ab28838, 1:100; Abcam, Cambridge, MA)antibodies and DAPI (Invitrogen). Tubes with cells that invaded collagenmatrices were observed using a LSM 710 confocal microscope (Carl Zeiss,Germany) and angiogenic activities were assessed according to numbers ofECs that had migrated to a depth >100 µm.

EC spheroid sprouting assaysThin Matrigel (BD Biosciences) matrices were formed by adding 10 µlof Matrigel to the bottom chambers of µ-Slide Angiogenesis plates(ibidi). Subsequently, detached ECs were inoculated at high density(4×104 cells/well) in HuMedia-EB2 medium supplemented 1% FBS andwere incubated until multicellular spheroids formed. After 24 h culture,spheroid cells were visualized using a ZEISS Axiovert A1 microscope (CarlZeiss) at 10× magnification, and the numbers of migrated cells per 20surface cells were counted on at least six spheroids.

Analyses of human JunB promoter activityAhuman JunB promoter region containing 2 kb of sequence upstream of thestart site was PCR amplified using genomic DNA from HMVECs and thefollowing primers: 5′-GAAGATCTAATTTCTGGCAGACATGTCT-3′and 5′-CCCAAGCTTCGCTGCGGTGACCGGACTGG-3′, and werecloned into pNL1.2 Nano-Luc-PEST vectors (Promega, Madison, WI).JunB promoter-Nano-Luc-PEST fragments were then cloned into pCDHvectors for lentivirus production. Subsequently, ECs were transduced withJunB promoter-Nano-Luc lentivirus and JunB promoter activity wasanalyzed by determining the NanoLuc luciferase activity using a NanoGlo system (Promega).

In utero microinjections into embryonic limbs of miceEC-tdTomato mouse embryos were accessed via the uterus under controlledanesthesia on E12.5. Abdominal cavities were then cut open with a scalpeland uterine horns were carefully extracted using ring forceps. Subsequently,2-µl lentiviral solutions (IFU=1.6×105 cells) were administered to forelimbsof embryos via in utero injections using 100-nm needles (AltairCorporation, Yokohama, Japan). The injection rate was controlled at0.2 µl/s using a syringe pump (YSP-201, YMC, Tokyo, Japan) that wascontrolled by a foot switch. After injection, uterine horns were carefullyreplaced in the abdominal cavities and muscle and skin incisions weresutured separately. Mice were then rested on a thermal support device andrecovery was monitored. At E17.5, embryos were harvested and limb skinvasculature was analyzed using whole-mount immunohistochemistry asdescribed below.

Whole-mount immunohistochemistryWhole-mount immunohistochemistry analyses of mouse limb skin fromembryoswas performed according to a previously described procedure (Li andMukouyama,2011)with somemodifications.Briefly,mouse limbs fromE9.5,E13.5 and E17.5 embryos were dissected and fixed in 4% paraformaldehyde(PFA) overnight with gentle agitation. After three washes with phosphate-buffered saline (PBS) containing 0.2% Triton X-100 (PBS-T), dermal tissueswere separated frommuscles and bones, and skin vasculature and neural fibersin dermal tissues were blocked with 10% goat serum in PBS-T, and were thenstained with anti-mouse-CD31 (MEC13.3, 1:200; BD Biosciences), anti-Neurofilament M (MAB1621, 1:200; Millipore), anti-JunB (ab28838 andab128878, 1:100 and 1:250, respectively; Abcam), eFluor-660-conjugated-anti-β tubulin class III (2G10-TB3, cat. no. 50-4510, 1:250 eBioscience, SanDiego, CA) and DAPI (Invitrogen). Alexa Fluor conjugates of anti-rat, anti-mouse, and anti-rabbit IgG (Invitrogen) were then used as secondaryantibodies and series of fluorescent Z-stack images of dermal tissuevasculature and nerve fiber networks were generated by using a LSM 710confocal microscope (Carl Zeiss).

Apoptosis assayAn apoptosis assay was performed using the Annexin-V ApoptosisDetection kit (BioVision, CA). ECs infected with the control shRNAvector or the shJunB vector, both containing a GFP marker, were detachedand re-suspended in binding buffer containing 5 µl of Annexin-V–biotinsolution and incubated for 5 min. After washing, the cells were incubatedwith avidin–APC. Annexin-V-positive and GFP-expressing cells wereanalyzed by confocal microscopy (Carl Zeiss).

Statistical and image analysesData are expressed as mean±s.d. or s.e.m. Statistical analyses were performedusing the R package (version 3.3.0; http://www.r-project.org). Pairwisedifferences between homoscedastic samples were identified using Student’st-tests, and those between heteroscedastic samples were identified usingWelch’s t-test. Image analysis and quantification were performed with the Fijipackage of ImageJ (http://fiji.sc/), and Z-stacks were flattened through amaximum intensity projections. Vasculature formation analyses wereperformed by counting numbers of neurovascular contacts and blood vesselbranches, and measuring branch lengths according to the criteria described inFig. S2. The images presented are representative of the analyzed data.

AcknowledgementsWe thank Kazumi Tanaka (Kanazawa Medical University) for her help andexperimental assistance.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsY. Yoshitomi and H.Y. conceived and designed the experiments; Y.Yoshitomi andH.Y. performed the majority of the experiments and analyzed the data with somecontributions from Y.I., T.I., H.S. and Y. Yoshitake. N.K. and T.H. assisted with DRGculture and in utero injection, respectively. Y. Yoshitomi and H.Y. interpreted thedata. Y. Yoshitomi and H.Y. wrote the manuscript.

FundingThis work was supported in part by the grants-in-aid from the Ministry of Education,Culture, Sports, Science and Technology (24790297 to Y.Y., 20590290 to H.Y.), andStrategic Research Project grants from the Kanazawa Medical University andMinistry of Education, Culture, Sports, Science and Technology, Japan (H2012-16[S1201022] to H.Y.).

Data availabilityThe JunB overexpression effect on microvascular endothelial cell culture microarraydata are available in Gene Expression Omnibus (GEO) under accession numberGSE93616.Microarray data for gene expression inmicrovascular endothelial cells co-cultured with dorsal root ganglion cells are available under accession numberGSE93696.

Supplementary informationSupplementary information available online athttp://jcs.biologists.org/lookup/doi/10.1242/jcs.196303.supplemental

925

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience

Page 11: JunB regulates angiogenesis and neurovascular parallel ...neurovascular juxtapositional alignment, the AP-1 transcription factor JunB was the most strongly and consistently upregulated

ReferencesBates, D., Taylor, G. I., Minichiello, J., Farlie, P., Cichowitz, A., Watson, N.,Klagsbrun, M., Mamluk, R. and Newgreen, D. F. (2003). Neurovascularcongruence results from a shared patterning mechanism that utilizesSemaphorin3A and Neuropilin-1. Dev. Biol. 255, 77-98.

Brooks, P. C., Clark, R. A. and Cheresh, D. A. (1994a). Requirement of vascularintegrin alpha v beta 3 for angiogenesis. Science 264, 569-571.

Brooks, P. C., Montgomery, A. M. P., Rosenfeld, M., Reisfeld, R. A., Hu, T., Klier,G. andCheresh, D. A. (1994b). Integrin alpha v beta 3 antagonists promote tumorregression by inducing apoptosis of angiogenic blood vessels. Cell 79,1157-1164.

Carmeliet, P. (2003). Blood vessels and nerves: common signals, pathways anddiseases. Nat. Rev. Genet. 4, 710-720.

Carmeliet, P. and Tessier-Lavigne, M. (2005). Common mechanisms of nerve andblood vessel wiring. Nature 436, 193-200.

Davis, G. E. and Senger, D. R. (2005). Endothelial extracellular matrix:biosynthesis, remodeling, and functions during vascular morphogenesis andneovessel stabilization. Circ. Res. 97, 1093-1107.

Desgrosellier, J. S. and Cheresh, D. A. (2010). Integrins in cancer: biologicalimplications and therapeutic opportunities. Nat. Rev. Cancer 10, 9-22.

Dorrell, M. I. and Friedlander, M. (2006). Mechanisms of endothelial cell guidanceand vascular patterning in the developing mouse retina. Prog. Retin. Eye Res. 25,277-295.

Estrach, S., Cailleteau, L., Franco, C. A., Gerhardt, H., Stefani, C., Lemichez, E.,Gagnoux-Palacios, L., Meneguzzi, G. and Mettouchi, A. (2011). Laminin-binding integrins induce Dll4 expression and Notch signaling in endothelial cells.Circ. Res. 109, 172-182.

Fellmann, C., Hoffmann, T., Sridhar, V., Hopfgartner, B., Muhar, M., Roth, M.,Lai, D. Y., Barbosa, I. A. M., Kwon, J. S., Guan, Y. et al. (2013). An optimizedmicroRNA backbone for effective single-copy RNAi. Cell Rep. 5, 1704-1713.

Gerhardt, H., Golding, M., Fruttiger, M., Ruhrberg, C., Lundkvist, A.,Abramsson, A., Jeltsch, M., Mitchell, C., Alitalo, K., Shima, D. et al. (2003).VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. CellBiol. 161, 1163-1177.

Guo, F.-J., Jiang, R., Xiong, Z., Xia, F., Li, M., Chen, L. and Liu, C.-J. (2014).IRE1a constitutes a negative feedback loop with BMP2 and acts as a novelmediator in modulating osteogenic differentiation. Cell Death Dis. 5, e1239.

Hayashi, H., Sano, H., Seo, S. and Kume, T. (2008). The Foxc2 transcription factorregulates angiogenesis via induction of integrin β3 expression. J. Biol. Chem. 283,23791-23800.

Kidoya, H., Naito, H., Muramatsu, F., Yamakawa, D., Jia, W., Ikawa, M., Sonobe,T., Tsuchimochi, H., Shirai, M., Adams, R. H. et al. (2015). APJ regulatesparallel alignment of arteries and veins in the skin. Dev. Cell 33, 247-259.

Kiesow, K., Bennewitz, K., Miranda, L. G., Stoll, S. J., Hartenstein, B., Angel, P.,Kroll, J. and Schorpp-Kistner, M. (2015). Junb controls lymphatic vasculardevelopment in zebrafish via miR-182. Sci. Rep. 5, 15007.

Kitagawa, M., Hojo, M., Imayoshi, I., Goto,M., Ando, M., Ohtsuka, T., Kageyama,R. and Miyamoto, S. (2013). Hes1 and Hes5 regulate vascular remodeling andarterial specification of endothelial cells in brain vascular development. Mech.Dev. 130, 458-466.

Klein, R. (2004). Eph/ephrin signaling in morphogenesis, neural development andplasticity. Curr. Opin. Cell Biol. 16, 580-589.

Kogata, N., Arai, Y., Pearson, J. T., Hashimoto, K., Hidaka, K., Koyama, T.,Somekawa, S., Nakaoka, Y., Ogawa, M., Adams, R. H. et al. (2006). Cardiacischemia activates vascular endothelial cadherin promoter in both preexistingvascular cells and bone marrow cells involved in neovascularization. Circ. Res.98, 897-904.

Koh, W., Stratman, A. N., Sacharidou, A. and Davis, G. E. (2008). In vitro threedimensional collagen matrix models of endothelial lumen formation duringvasculogenesis and angiogenesis. Methods Enzymol. 443, 83-101.

Leu, S.-J., Lam, S. C.-T. and Lau, L. F. (2002). Pro-angiogenic activities of CYR61(CCN1) mediated through integrins alphavbeta3 and alpha6beta1 in humanumbilical vein endothelial cells. J. Biol. Chem. 277, 46248-46255.

Li, W. and Mukouyama, Y. S. (2011). Whole-mount immunohistochemical analysisfor embryonic limb skin vasculature: a model system to study vascular branchingmorphogenesis in embryo. J. Vis. Exp. 51, 2620.

Li, W., Kohara, H., Uchida, Y., James, J. M., Soneji, K., Cronshaw, D. G., Zou, Y.-R., Nagasawa, T. and Mukouyama, Y.-S. (2013). Peripheral nerve-derivedCXCL12 and VEGF-A regulate the patterning of arterial vessel branching indeveloping limb skin. Dev. Cell 24, 359-371.

Madisen, L., Zwingman, T. A., Sunkin, S. M., Oh, S. W., Zariwala, H. A., Gu, H.,Ng, L. L., Palmiter, R. D., Hawrylycz, M. J., Jones, A. R. et al. (2010). A robustand high-throughput Cre reporting and characterization system for the wholemouse brain. Nat. Neurosci. 13, 133-140.

Mukouyama, Y.-S., Shin, D., Britsch, S., Taniguchi, M. and Anderson, D. J.(2002). Sensory nerves determine the pattern of arterial differentiation and bloodvessel branching in the skin. Cell 109, 693-705.

Mukouyama, Y.-S., Gerber, H.-P., Ferrara, N., Gu, C. and Anderson, D. J. (2005).Peripheral nerve-derived VEGF promotes arterial differentiation via neuropilin 1-mediated positive feedback. Development 132, 941-952.

Pasquale, E. B. (2005). Eph receptor signalling casts a wide net on cell behaviour.Nat. Rev. Mol. Cell Biol. 6, 462-475.

Plow, E. F., Haas, T. A., Zhang, L., Loftus, J. and Smith, J. W. (2000). Ligandbinding to integrins. J. Biol. Chem. 275, 21785-21788.

Poliakov, A., Cotrina, M. and Wilkinson, D. G. (2004). Diverse roles of ephreceptors and ephrins in the regulation of cell migration and tissue assembly.Dev.Cell 7, 465-480.

Potente, M., Urbich, C., Sasaki, K.-I., Hofmann, W. K., Heeschen, C., Aicher, A.,Kollipara, R., DePinho, R. A., Zeiher, A. M. and Dimmeler, S. (2005).Involvement of Foxo transcription factors in angiogenesis and postnatalneovascularization. J. Clin. Invest. 115, 2382-2392.

Raffaello, A., Milan, G., Masiero, E., Carnio, S., Lee, D., Lanfranchi, G.,Goldberg, A. L. and Sandri, M. (2010). JunB transcription factor maintainsskeletal muscle mass and promotes hypertrophy. J. Cell Biol. 191, 101-113.

Ruhrberg, C., Gerhardt, H., Golding, M.,Watson, R., Ioannidou, S., Fujisawa, H.,Betsholtz, C. and Shima, D. T. (2002). Spatially restricted patterning cuesprovided by heparin-binding VEGF-A control blood vessel branchingmorphogenesis. Genes Dev. 16, 2684-2698.

Santaguida, M., Schepers, K., King, B., Sabnis, A. J., Forsberg, E. C., Attema,J. L., Braun, B. S. and Passegue, E. (2009). JunB protects against myeloidmalignancies by limiting hematopoietic stem cell proliferation and differentiationwithout affecting self-renewal. Cancer Cell 15, 341-352.

Schorpp-Kistner, M., Wang, Z.-Q., Angel, P. and Wagner, E. F. (1999). JunB isessential for mammalian placentation. EMBO J. 18, 934-948.

Somanath, P. R., Malinin, N. L. and Byzova, T. V. (2009). Cooperation betweenintegrin alphavbeta3 and VEGFR2 in angiogenesis. Angiogenesis 12, 177-185.

Son, J.-S., Sahoo, A., Chae, C.-S., Hwang, J.-S., Park, Z. Y. and Im, S.-H. (2011).JunB and c-Rel cooperatively enhance Foxp3 expression during inducedregulatory T cell differentiation. Biochem. Biophys. Res. Commun. 407, 141-147.

Stenzel, D., Franco, C. A., Estrach, S., Mettouchi, A., Sauvaget, D., Rosewell, I.,Schertel, A., Armer, H., Domogatskaya, A., Rodin, S. et al. (2011). Endothelialbasement membrane limits tip cell formation by inducing Dll4/Notch signalling invivo. EMBO Rep. 12, 1135-1143.

van Dam, H. and Castellazzi, M. (2001). Distinct roles of Jun:Fos and Jun:ATFdimers in oncogenesis. Oncogene 20, 2453-2464.

Walchli, T., Wacker, A., Frei, K., Regli, L., Schwab, M. E., Hoerstrup, S. P.,Gerhardt, H. and Engelhardt, B. (2015). Wiring the vascular network with neuralcues: A CNS perspective. Neuron 87, 271-296.

926

RESEARCH ARTICLE Journal of Cell Science (2017) 130, 916-926 doi:10.1242/jcs.196303

Journal

ofCe

llScience