Skip to main content
Erschienen in: Vascular Cell 1/2013

Open Access 01.12.2013 | Review

The importance of microglia in the development of the vasculature in the central nervous system

verfasst von: Tom Arnold, Christer Betsholtz

Erschienen in: Vascular Cell | Ausgabe 1/2013

Abstract

The body’s vascular system is thought to have developed in order to supply oxygen and nutrients to cells beyond the reach of simple diffusion. Hence, relative hypoxia in the growing central nervous system (CNS) is a major driving force for the ingression and refinement of the complex vascular bed that serves it. However, even before the establishment of this CNS vascular system, CNS-specific macrophages (microglia) migrate into the brain. Recent studies in mice point to the fundamental importance of microglia in shaping CNS vasculature during development, and re-shaping these vessels during pathological insults. In this review, we discuss the origin of CNS microglia and their localization within the brain based on data obtained in mice. We then review evidence supporting a functional role of these microglia in developmental angiogenesis. Although pathologic processes such as CNS ischemia may subvert the developmental functions of microglia/macrophages with significant effects on brain neo-angiogenesis, we have left this topic to other recent reviews (Nat Rev Immunol 9:259–270, 2009 and Trends Mol Med 17:743–752, 2011).
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​2045-824X-5-4) contains supplementary material, which is available to authorized users.
An erratum to this article is available at http://​dx.​doi.​org/​10.​1186/​2045-824X-5-12.

Competing interests

The authors declare that they have no competing interests.

Microglia – specialized macrophages of the CNS

Microglia are specialized macrophages of the central nervous system involved in immune regulation, tissue development, homeostasis and wound repair. Microglia were first observed by Virchow in the mid-nineteenth century (see), and described in greater detail by Pio del Rio-Hortega in 1932. In this almost prescient work, del Rio-Hortega described microglia morphology, plasticity during development and with pathological insult, their cellular origin, and microglia association with white matter tracts and blood vessels. Despite an immense amount of research on microglia origin and function since then, these early views remain surprisingly accurate.

Microglia derive from primitive yolk sac macrophages

Microglia belong to the mononuclear phagocytic system - a family of cells that includes committed precursors in the bone marrow, circulating blood monocytes and tissue macrophages in every organ of the body including the CNS. Mononuclear phagocytes are typified by their ability to ingest large particles; their morphology; their expression of common surface markers including CD11b, CD68, Colony Stimulating Factor 1 Receptor (CSF1R), chemokine receptor CXCR3, and plasma membrane glycoprotein F4/80 [1]; and their presumed hematopoietic origin. While microglia certainly meet the functional and morphological definition of a mononuclear phagocyte [24], their developmental origin has until recently been less clear.
In mice, hematopoietic stem cells (HSCs) emerge from the dorsal aorto-gonado-mesonephros (AGM) region 10.5 days after conception (embryonic day (E) 10.5), then migrate to the fetal liver where they expand and differentiate before definitive hematopoiesis in the spleen and bone marrow [58]. In adult mice, blood monocytes, classical dendritic cells, and certain tissue macrophages derive from, and are continuously replaced by, bone marrow-derived HSCs. It was previously thought that microglia arose from hematopoietic precursors in two waves of recruitment and differentiation [9, 10]. However, it is now clear, based on evidence from bird, fish and mammals, that yolk-sac derived macrophage precursors contribute significantly, if not entirely, to the brain’s microglia. In avian embryos, analyses using chick-quail transplantation and parabiosis chimeras show that yolk sac-derived macrophages migrate to and invade the CNS through the pial basal lamina before and independent of CNS vacularization [11, 12]. Subsequent live recordings of cell movements in zebrafish embryos revealed that yolk sac-derived macrophages migrate through the cephalic mesenchyme before its vascularization to reach the brain pial surface and the roof of the 4th ventricle, from where they subsequently invade the neuroepithelium and eventually acquire microglial characteristics [13]. Recently, fate mapping studies in the mouse using genetic lines such as Cx3cr1-green fluorescent protein (GFP), Csf1r-GFP, and cell surface markers, show that microglial precursors originate in the yolk-sac, colonize the surface of the brain between E9.5 and E10.5 (i.e. before the onset of HSC formation in the AGM region), then appear within the brain neuroepithelium in association with blood vessels by E10.5 (Figure 1) [1417]. Lineage analysis with tamoxifen-inducible Runx1-CreERtm[14] or Csf1r-CreERtm mice [15] suggests that the first wave of yolk-sac derived microglia is specified before E8.0. Microglia proliferate throughout embryogenesis and self-renew without significant contribution from the bone marrow in the steady state [14, 15, 18]. While bone-marrow derived monocytes may infiltrate the brain parenchyma in conjunction with irradiation or inflammation [19], these cells later disappear, and do not significantly contribute to the population of resident microglia [18].
The yolk sac origin of microglia is further supported by elegant experiments done in mice lacking the transcription factors PU.1 [15, 2022] or Myb [15], and in mice with inactivated Csf1 (CSF1 op/op mice) [23], or Csf1r genes [24, 25]. PU.1 acts in part by activating transcription of Csf1r[26], which is highly expressed in macrophages and microglia during development and throughout adult life [14]. The major ligands of CSF1R, IL34 and CSF1, are highly expressed in the brain parenchyma during microglial colonization [27]. Mice with genetic deletion of Pu.1[15] or Csf1r[14] have substantially reduced yolk sac macrophages and microglia, but normal numbers of circulating monocytes [14, 15]. Csf1 op/op mice have a milder reduction in microglia, consistent with an important role for IL34 in microglial homeostasis [24, 27]. Further work with Myb-deficient mice clarified the distinct lineage of microglia. Genetic loss of Myb blocks the generation of HSCs and their progeny (including circulating monocytes and granulocytes), but these mice have normal numbers of tissue macrophages and microglial cells [15]. Similarly, myeloid-specific expression of diphtheria toxin in LysMCre;Rosa26DTA transgenic mice eliminates monocyte-derived macrophages without effect on resident microglia [16]. Taken together, these studies indicate that the large majority of embryonic and adult brain microglia are derived from early yolk sac precursors.

Patterns of brain colonization by microglia

Once born, yolk-sac derived macrophage precursors migrate into and colonize the whole (mouse) embryo between E9.5 and E10.5 (Figure 1) [14, 15]. The first organ to be colonized is the brain. Subsequent phases of microglial brain colonization follow a stereotyped pattern (see [28]). Microglia invade the brain through the pial surface, then migrate and proliferate, populating the brain in a dorsal-to-ventral and rostral-to-caudal gradient. During this time, microglia associate with radial glia and blood vessels, and are found in close proximity to dying cells. Eventually, microglia are notably excluded from the neuroepithelium and cortical plate, and then are widely distributed in the adult brain, except in areas of densely packed neuron cell bodies such as the pyramidal cell layer. The early association of microglia around blood vessels has led to the hypothesis that microglia may enter the brain through the developing vasculature. Indeed, E10.5 Ncx1 −/− embryos [29], which lack heartbeat and blood flow, are devoid of microglia [14]. However, it is not clear whether lack of blood flow per se disrupts brain vascular development in these mutants (they may lack brain vasculature altogether). It is possible that some microglia migrate along the abluminal surface of pial penetrating vessels, independent of blood flow, or that a small population of microglia (not detected by flow cytometry in [14] initially invades the brain without contact with vessels [16, 30]. Notably, microglia populate the embryonic retina well before post-natal retinal angiogenesis occurs [16, 31, 32], and as mentioned above appear to colonize zebrafish and chick brains before CNS vascular invasion [1113]. Shortly after entering the neuroepithelium, microglia associate with the developing blood vessels in the brain and retina [1113, 17, 33], and microglia and vessels may therefore directly influence each other’s development, an idea that lends support from ex vivo studies [32].

Microglia shape the developing CNS vasculature

Because the developing CNS lacks intrinsic vasculature, CNS blood vessel development occurs exclusively via angiogenesis [3436]. Attracted by proangiogenic signals, new capillaries sprout from perineural vessels, and invade the neuroectoderm around E10 in mice. These nascent capillaries are composed of tip cells at the vascular front, followed by proliferative stalk cells. Tip cells extend filopodia toward guidance cues such as VEGF-A. VEGF-A induces expression of the Notch ligand, Dll4, predominately in tip cells. Dll4 then activates Notch in adjacent cells, which down-regulates VEGF receptors and up-regulates angio-suppressive factors like sFlt1 and Jagged-1, promoting a stalk cell phenotype (reviewed in [36]). The interplay between VEGF and Notch signaling is highly regulated with additional inputs from other major signaling pathways including BMPs [37, 38], Semaphorins [39], and Wnt/βcatenin [40, 41]. Additional signaling pathways that regulate tip cell formation and sprouting include sphingosine-1-phosphate and its receptor S1pr1 [4244]. During vascular sprouting, tip cells anastomose with neighboring tip cells, creating vascular loops. In this way, vessels sprout, extend, branch and anastomose, iteratively, toward the center of the neural tube where they establish a temporary plexus, termed the periventricular vascular plexus (PVP), around the CNS ventricular spaces and spinal cord’s central canal [4547]. As the CNS grows and differentiates, these vessels associate with microglia, pericytes, neuroepithelial radial glia and neuroblasts, and later astrocytes; CNS vessels are refined, arteries and veins are established, and the mature neurovascular system takes form.
The retina and optic nerve represent highly specialized extensions of the forebrain. While its vascularization occurs by angiogenic sprouting similar to the brain, the timing and scaffolds that guide angiogenesis are partly different [48]. During the first week of life, an astrocytic network arising from the optic nerve invades into the retina in a centrifugal fashion. As the primitive hyaloid (hv) vessels that supported the embryonic eye development regress, a new primary vascular plexus extends into the retina, following structural and morphogenic cues provided by astrocytes and Müller glia. At 7 to 9 days of postnatal life, vessels sprout perpendicularly into deeper layers of the retina, forming a deep vascular plexus in the outer plexiform retinal layer (OPL). During the next 3 weeks, retinal vasculature continues to sprout, remodel and differentiate into arteries and veins and a mature neurovascular network is established by 6 weeks of age.

Microglia influence CNS vascular development

As discussed above, microglia migrate into the CNS and retinal neuroepithelium before vessels do. Microglia are therefore uniquely positioned to influence the early sprouting, migration, anastomosis, and refinement of the growing CNS and retinal vascular systems. Studies of angiogenesis after microglial depletion, or in mice lacking microglia, strongly support this concept. Checchin et al. [49] administered clodronate liposomes either systemically to deplete macrophages and circulating monocytes, or intravitreally, which depleted retinal microglia without reducing circulating monocytes. These authors found that reducing retinal microglia numbers was associated with a decrease in retinal vascular density. Selective depletion of circulating monocytes had no impact on retinal blood vessels. Importantly, intravitreal co-administration of microglia with clodronate restored vascularity of the developing retina, suggesting a microglia-specific effect on retinal vascular development. Similarly, Kubota et al. [33] found that Csf1 op/op mice, which initially lack retinal microglia, have a significant decrease in branching of the primary vascular plexus. Also, intra-vitreal administration of CSF1-neutralizing antibodies, or systemic administration of a CSF1R kinase inhibitor, decreased microglia numbers with commensurate reductions in vascular branching. Branching in these mice recovered as development progressed, suggesting that microglia principally effect developmental vascular remodeling, but do not contribute to maintenance of adult vascular patterns. Interestingly, they found that Csf1 op/op mice, and mice with pharmacologic microglial depletion, have comparatively normal numbers of endothelial tip cells and filopodia. This suggests that microglia facilitate branching anastomosis, but not tip cell extension. In contrast, Unoki et al [50] found that depletion of microglia using clodronate liposomes increased VEGF-mediated neovascular sprouts in an ex vivo retina culture model.
The concept that microglia may act to “bridge” vascular sprouts during CNS vascular development was introduced by the work of Fantin et al. [16]. This group studied vascular ingression and branching in embryonic hindbrains and retinas of mice lacking macrophages and microglia (Pu.1 −/− and Csf1 op/op mice). In the hindbrain, microglia numbers were correlated with numbers of branch points, and were frequently found to be in contact with neighboring endothelial sprouts. Loss of microglia was associated with a significant decrease in the numbers of vascular branch points in the brain periventricular vascular plexus, without significant effects on the numbers of tip cells, filopodia, or radial vessels ingressing from the surface of the brain (see also Figure 1). Selective depletion of circulating monocytes using LysmCre;Rosa26DTA transgenic mice did not affect hindbrain vascular branching, again suggesting that resident microglia are principally responsible for facilitated branching. As VEGF is central to tip cell guidance and vascular branching, and VEGF functions as a macrophage chemoattractant in tumors and in vitro, the authors evaluated VEGF mRNA levels in the hindbrains of microglia-deficient embryos and found no difference. They also analyzed microglia numbers and branching patterns in mice lacking VEGF, or with selective expression of VEGF120 (vegfa 120/120 mice, which lack heparin binding VEGF isoforms while retaining the diffusible VEGF120 isoform). These mutant mice revealed abnormalities in hindbrain angiogenesis distinct from those observed in microglia-deficient mice: vegfa 120/120 mice had more global vascular deficiencies, including decreases in the numbers of tip cells and penetrating radial vessels, and vegfa 120/120 and vegfa flox/+ ;NesCre mice had a more pronounced reduction in vascular branching than microglia-deficient mice. Microglia numbers were unaffected in these VEGF mutants. These results imply that microglia are not a significant source of VEGF, that VEGF (unlike CSF1 or IL34) is not a major chemoattractant or survival cue for microglia, and that the mechanisms of microglia-facilitated branching may be distinct from, and complimentary to, VEGF-mediated sprouting.
A recent report from our laboratory confirmed and extended many of these observations [32]. We found that microglia-deficient mice (Csf1 op/op mutants and Pu.1 knockouts) had reduced numbers of vascular branch points in the retina, and that the angle of filopodia extending from tip cells was reduced in these mice. We used microglia co-cultured in collagen matrix with mouse aortic rings to study microglia-vascular interactions more deeply. These studies revealed an apparent two-way communication between aortic rings and microglia: aortic rings induced the migration of microglia towards the ring, while microglia significantly increased vascular branches emanating from the ring. Interestingly, media from cultured microglia added separately to cultured aortic rings had a similar, albeit less potent, effect on vascular branching. This suggests microglia-blood vessel contact enhances, but is not necessary to induce branching, and that microglia may release soluble factors that stimulate sprouting/branching. Addition of soluble Flt1 (a VEGFR1 ectodomain that traps and neutralizes VEGFA, VEGFB and, placenta growth factor (PIGF) or VEGFR1 neutralizing antibody to microglia-aortic ring cultures did not effectively inhibit microglia-induced branching, suggesting that VEGFA and sFlt1 are not major microglia-derived factors responsible for branching induction.
In contrast to these findings, Stefater et al. [51] recently uncovered a mechanism whereby microglia may suppress angiogenic branching through a Wnt-Flt1 pathway. Here, they found that microglia associated with the deep retinal vascular plexus specifically express various Wnt signaling components including Wnt5a and Wnt11 ligands, and Wnt receptors Fzd7, Fzd8 and Lrp5. Microglia-specific haploinsufficiency of the common Wnt-ligand transporter, Wls (Wls flox/+ ;Csf1rCre mice), resulted in increased vascular branching in the deep vascular plexus. Similarly, Wnt5a and Wnt11 haploinsufficiency resulted in a similar phenotype. Interestingly, deletion of Lrp5 from microglia had the opposite effect, with a significant reduction in vascular branching. They found that Wnt5a induced expression of sFlt1, that Wls flox/+ ;Csf1rCre mice have reduced expression of sFlt1, and that microglia-specific haploinsufficiency of Flt1 phenocopies the Wls and Wnt5a and Wnt11 mutants. Taken together, their results indicate that microglia can suppress vascular branching in the deep retinal vascular plexus by secreting Wnt ligands, which induce, in an autocrine fashion, secretion of the VEGF inhibitory protein, sFlt1. These results appear to contrast with those reported by Fantin et al., who found no difference in branch density in the deep retinal vascular plexus in the absence of microglia [13], which they explained was the net outcome of less branching and less pruning. This result pinpoints the potential complex role of microglial cells in retinal angiogenesis, as regulators of endothelial sprouting, branch fusion and regression.
The feedback loop between VEGF and Notch involves regulation of both VEGFR-2 and VEGFR-3, although the individual contribution of each of these VEGF receptors remains unclear [41, 5254]. The primary ligands for VEGFR-3, VEGF-C and VEGF-D, are highly expressed by microglia, and VEGF-C-positive microglia are found near the fusion points of VEGFR-3-positive vascular sprouts [54]. While Vegfd knockout mice have no apparent retinal vascular phenotype, Vegfc heterozygous mice display delayed retinal vascularization and decreased branching, but increased vessel sprouting and filopodia [54]. Interestingly, this phenotype is different from mice with loss of function deletions or antibody blockade of VEGFR-3 [52, 53]. There are alternative explanations for these discrepancies. One group proposes that VEGFR-3 has both ligand-dependent (and pro-angiogenic) and ligand-independent (anti-angiogenic) signaling activities [54]. Another group suggests that ligand-independent signaling by VEGFR-3 is pro-angiogenic (when Notch signaling is suppressed) [52]. Further experimentation should clarify the roles of microglia-derived VEGF-C/D-VEGFR3 signaling in vascular development.
Other groups have recently explored potential roles for Notch signaling in microglia-endothelial cell interactions. Outtz et al. [55] found that Notch signaling is activated in retinal microglia, which are closely associated with endothelial tip cells expressing the Notch ligand Dll4. Moreover, genetic deletion of Notch1 in retinal microglia led to a subtle reduction in the numbers of microglia found at the vascular front. Interestingly, Hoffman et al. [56] found that the Notch ligand, Jagged1, is highly expressed in perivascular cells of the retina, including microglia. Further studies should evaluate the specific roles of Notch signaling in microglia, and the impact of this signaling on retinal vascular development.

Outlook

The development of organ-specific vascular beds is dependent upon the close communication between the vascular cells (endothelial cells and mural cells) on the one hand, and resident cells of the organ, on the other. The CNS vasculature is in many ways unique in its anatomy and regulation, and it harbors a highly specific barrier – the blood–brain, or blood-retina, barrier. The development of the CNS vasculature hence occurs in tight association with the development of other components of the CNS, and as a result of reciprocal communication between the endothelial cells and different emerging CNS cell types. This review focuses on the role of microglial cells – a CNS-specific type of macrophage – and also mentions in passing the importance of other cells types, such as radial glial cells and astrocytes, as sources of VEGFs, Wnts and other signaling molecules that control the shape and function of the emerging vasculature. Although recent studies provide compelling evidence for a role of microglia in shaping the nascent vascular plexuses in the brain and retina, the molecular mechanisms remain to be elucidated, and the possibility remains that microglia play a different role at different locations, for example in the different retinal capillary plexuses. It also remains to be shown what role, if any, microglia play in vascular homoeostasis in adult physiological and pathophysiological processes. Microglial cells become activated in conjunction with pathological insults and disruption of the blood–brain barrier, where they may play protective or pathogenic roles. These data are not discussed in the present review, which is focused on physiological development. However, the awareness of microglia as a unique CNS cell type with a distinct ontogeny and equipped with specific developmental and pathological functions compared to other glial cell types, has recently increased. Our perspectives on these cells will undoubtedly grow rapidly in the coming years.

Ethical approval

Animal experiments were approved by the Stockholm’s North Ethical Committee for Animal Research.

Acknowledgments

This work has been supported by a Leducq Foundation Transatlantic Network of Excellence and a Transatlantic Career Development Award. Pu.1−/− knockout embryos generously supplied by Suzanne Abdelazim and Yosuke Mukoyama, Ph.D., Laboratory of Stem Cell and Neuro-Vascular Biology Genetics and Developmental Biology Center National Heart, Lung, and Blood Institute (NHLBI).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Kettenmann H, Hanisch U-K, Noda M, Verkhratsky A: Physiology of microglia. Physiol Rev. 2011, 91: 461-553. 10.1152/physrev.00011.2010.CrossRefPubMed Kettenmann H, Hanisch U-K, Noda M, Verkhratsky A: Physiology of microglia. Physiol Rev. 2011, 91: 461-553. 10.1152/physrev.00011.2010.CrossRefPubMed
2.
Zurück zum Zitat Van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL: The mononuclear phagocyte system: A new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ. 1972, 46: 845-PubMedCentralPubMed Van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL: The mononuclear phagocyte system: A new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ. 1972, 46: 845-PubMedCentralPubMed
4.
Zurück zum Zitat Hanisch U-K, Kettenmann H: Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007, 10: 1387-1394. 10.1038/nn1997.CrossRefPubMed Hanisch U-K, Kettenmann H: Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007, 10: 1387-1394. 10.1038/nn1997.CrossRefPubMed
5.
Zurück zum Zitat Peri F, Nüsslein-Volhard C: Live imaging of neuronal degradation by microglia reveals a role for v0-atpase a1 in phagosomal fusion in vivo. Cell. 2008, 133: 916-927. 10.1016/j.cell.2008.04.037.CrossRefPubMed Peri F, Nüsslein-Volhard C: Live imaging of neuronal degradation by microglia reveals a role for v0-atpase a1 in phagosomal fusion in vivo. Cell. 2008, 133: 916-927. 10.1016/j.cell.2008.04.037.CrossRefPubMed
6.
Zurück zum Zitat Sieger D, Moritz C, Ziegenhals T, Prykhozhij S, Peri F: Long-range ca2+ waves transmit brain-damage signals to microglia. Dev Cell. 2012, 22: 1138-1148. 10.1016/j.devcel.2012.04.012.CrossRefPubMed Sieger D, Moritz C, Ziegenhals T, Prykhozhij S, Peri F: Long-range ca2+ waves transmit brain-damage signals to microglia. Dev Cell. 2012, 22: 1138-1148. 10.1016/j.devcel.2012.04.012.CrossRefPubMed
7.
Zurück zum Zitat Samokhvalov IM, Samokhvalova NI, Nishikawa S-I: Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature. 2007, 446: 1056-1061. 10.1038/nature05725.CrossRefPubMed Samokhvalov IM, Samokhvalova NI, Nishikawa S-I: Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature. 2007, 446: 1056-1061. 10.1038/nature05725.CrossRefPubMed
8.
Zurück zum Zitat Kissa K, Herbomel P: Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature. 2010, 464: 112-115. 10.1038/nature08761.CrossRefPubMed Kissa K, Herbomel P: Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature. 2010, 464: 112-115. 10.1038/nature08761.CrossRefPubMed
9.
Zurück zum Zitat Boisset J-C, van Cappellen W, Andrieu-Soler C, Galjart N, Dzierzak E, Robin C: In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature. 2010, 464: 116-120. 10.1038/nature08764.CrossRefPubMed Boisset J-C, van Cappellen W, Andrieu-Soler C, Galjart N, Dzierzak E, Robin C: In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature. 2010, 464: 116-120. 10.1038/nature08764.CrossRefPubMed
10.
Zurück zum Zitat Bertrand JY, Chi NC, Santoso B, Teng S, Stainier DYR, Traver D: Haematopoietic stem cells derive directly from aortic endothelium during development. Nature. 2010, 464: 108-111. 10.1038/nature08738.PubMedCentralCrossRefPubMed Bertrand JY, Chi NC, Santoso B, Teng S, Stainier DYR, Traver D: Haematopoietic stem cells derive directly from aortic endothelium during development. Nature. 2010, 464: 108-111. 10.1038/nature08738.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Perry VH, Hume DA, Gordon S: Immunohistochemical localization of macrophages and microglia in the adult and developing mouse brain. Neuroscience. 1985, 15: 313-326. 10.1016/0306-4522(85)90215-5.CrossRefPubMed Perry VH, Hume DA, Gordon S: Immunohistochemical localization of macrophages and microglia in the adult and developing mouse brain. Neuroscience. 1985, 15: 313-326. 10.1016/0306-4522(85)90215-5.CrossRefPubMed
12.
Zurück zum Zitat Chan WY, Kohsaka S, Rezaie P: The origin and cell lineage of microglia: New concepts. Brain Res Rev. 2007, 53: 344-354. 10.1016/j.brainresrev.2006.11.002.CrossRefPubMed Chan WY, Kohsaka S, Rezaie P: The origin and cell lineage of microglia: New concepts. Brain Res Rev. 2007, 53: 344-354. 10.1016/j.brainresrev.2006.11.002.CrossRefPubMed
13.
Zurück zum Zitat Kurz H, Christ B: Embryonic CNS macrophages and microglia do not stem from circulating, but from extravascular precursors. GLIA. 1998, 22: 98-102. 10.1002/(SICI)1098-1136(199801)22:1<98::AID-GLIA10>3.0.CO;2-V.CrossRefPubMed Kurz H, Christ B: Embryonic CNS macrophages and microglia do not stem from circulating, but from extravascular precursors. GLIA. 1998, 22: 98-102. 10.1002/(SICI)1098-1136(199801)22:1<98::AID-GLIA10>3.0.CO;2-V.CrossRefPubMed
14.
Zurück zum Zitat Cuadros MA, Martin C, Coltey P, Almendros A, Navascués J: First appearance, distribution, and origin of macrophages in the early development of the avian central nervous system. J Comp Neurol. 1993, 330: 113-129. 10.1002/cne.903300110.CrossRefPubMed Cuadros MA, Martin C, Coltey P, Almendros A, Navascués J: First appearance, distribution, and origin of macrophages in the early development of the avian central nervous system. J Comp Neurol. 1993, 330: 113-129. 10.1002/cne.903300110.CrossRefPubMed
15.
Zurück zum Zitat Herbomel P, Thisse B, Thisse C: Zebrafish early macrophages colonize cephalic mesenchyme and developing brain, retina, and epidermis through a M-CSF receptor-dependent invasive process. Dev Biol. 2001, 238: 274-288. 10.1006/dbio.2001.0393.CrossRefPubMed Herbomel P, Thisse B, Thisse C: Zebrafish early macrophages colonize cephalic mesenchyme and developing brain, retina, and epidermis through a M-CSF receptor-dependent invasive process. Dev Biol. 2001, 238: 274-288. 10.1006/dbio.2001.0393.CrossRefPubMed
16.
Zurück zum Zitat Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S: Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010, 330: 841-845. 10.1126/science.1194637.PubMedCentralCrossRefPubMed Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S: Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010, 330: 841-845. 10.1126/science.1194637.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K: A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. 2012, 336: 86-90. 10.1126/science.1219179.CrossRefPubMed Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K: A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. 2012, 336: 86-90. 10.1126/science.1219179.CrossRefPubMed
18.
Zurück zum Zitat Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S: Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of vegf-mediated endothelial tip cell induction. Blood. 2010, 116 (5): 829-840. 10.1182/blood-2009-12-257832.PubMedCentralCrossRefPubMed Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S: Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of vegf-mediated endothelial tip cell induction. Blood. 2010, 116 (5): 829-840. 10.1182/blood-2009-12-257832.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Ovchinnikov DA, van Zuylen WJM, DeBats CEE, Alexander KA, Kellie S, Hume DA: Expression of gal4-dependent transgenes in cells of the mononuclear phagocyte system labeled with enhanced cyan fluorescent protein using csf1r-gal4vp16/UAS-ECFP double-transgenic mice. J Leukoc Biol. 2008, 83: 430-433.CrossRefPubMed Ovchinnikov DA, van Zuylen WJM, DeBats CEE, Alexander KA, Kellie S, Hume DA: Expression of gal4-dependent transgenes in cells of the mononuclear phagocyte system labeled with enhanced cyan fluorescent protein using csf1r-gal4vp16/UAS-ECFP double-transgenic mice. J Leukoc Biol. 2008, 83: 430-433.CrossRefPubMed
20.
Zurück zum Zitat Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FMV: Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 2011, 14: 1142-1149. 10.1038/nn.2887.CrossRefPubMed Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FMV: Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 2011, 14: 1142-1149. 10.1038/nn.2887.CrossRefPubMed
21.
Zurück zum Zitat Ransohoff RM: Microglia and monocytes: Tis plain the twain meet in the brain. Nat Neurosci. 2011, 14: 1098-1100. 10.1038/nn.2917.CrossRefPubMed Ransohoff RM: Microglia and monocytes: Tis plain the twain meet in the brain. Nat Neurosci. 2011, 14: 1098-1100. 10.1038/nn.2917.CrossRefPubMed
22.
Zurück zum Zitat Dakic A, Metcalf D, Di Rago L, Mifsud S, Wu L, Nutt SL: PU.1 regulates the commitment of adult hematopoietic progenitors and restricts granulopoiesis. J Exp Med. 2005, 201: 1487-1502. 10.1084/jem.20050075.PubMedCentralCrossRefPubMed Dakic A, Metcalf D, Di Rago L, Mifsud S, Wu L, Nutt SL: PU.1 regulates the commitment of adult hematopoietic progenitors and restricts granulopoiesis. J Exp Med. 2005, 201: 1487-1502. 10.1084/jem.20050075.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, Baribault H: Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 1996, 15: 5647-5658.PubMedCentralPubMed McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, Baribault H: Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 1996, 15: 5647-5658.PubMedCentralPubMed
24.
Zurück zum Zitat Nerlov C, Graf T: PU.1 induces myeloid lineage commitment in multipotent hematopoietic progenitors. Genes Dev. 1998, 12: 2403-2412. 10.1101/gad.12.15.2403.PubMedCentralCrossRefPubMed Nerlov C, Graf T: PU.1 induces myeloid lineage commitment in multipotent hematopoietic progenitors. Genes Dev. 1998, 12: 2403-2412. 10.1101/gad.12.15.2403.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Blevins G, Fedoroff S: Microglia in colony-stimulating factor 1-deficient op/op mice. J Neurosci Res. 1995, 40: 535-544. 10.1002/jnr.490400412.CrossRefPubMed Blevins G, Fedoroff S: Microglia in colony-stimulating factor 1-deficient op/op mice. J Neurosci Res. 1995, 40: 535-544. 10.1002/jnr.490400412.CrossRefPubMed
26.
Zurück zum Zitat Erblich B, Zhu L, Etgen AM, Dobrenis K, Pollard JW: Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS One. 2011, 6: e26317-10.1371/journal.pone.0026317.PubMedCentralCrossRefPubMed Erblich B, Zhu L, Etgen AM, Dobrenis K, Pollard JW: Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS One. 2011, 6: e26317-10.1371/journal.pone.0026317.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Chitu V, Stanley ER: Colony-stimulating factor-1 in immunity and inflammation. Curr Opin Immunol. 2006, 18: 39-48. 10.1016/j.coi.2005.11.006.CrossRefPubMed Chitu V, Stanley ER: Colony-stimulating factor-1 in immunity and inflammation. Curr Opin Immunol. 2006, 18: 39-48. 10.1016/j.coi.2005.11.006.CrossRefPubMed
28.
Zurück zum Zitat Reddy MA, Yang BS, Yue X, Barnett CJ, Ross IL, Sweet MJ: Opposing actions of c-ets/PU.1 and c-myb protooncogene products in regulating the macrophage-specific promoters of the human and mouse colony-stimulating factor-1 receptor (c-fms) genes. J Exp Med. 1994, 180: 2309-2319. 10.1084/jem.180.6.2309.CrossRefPubMed Reddy MA, Yang BS, Yue X, Barnett CJ, Ross IL, Sweet MJ: Opposing actions of c-ets/PU.1 and c-myb protooncogene products in regulating the macrophage-specific promoters of the human and mouse colony-stimulating factor-1 receptor (c-fms) genes. J Exp Med. 1994, 180: 2309-2319. 10.1084/jem.180.6.2309.CrossRefPubMed
29.
Zurück zum Zitat Wei S, Nandi S, Chitu V, Yeung Y-G, Yu W, Huang M: Functional overlap but differential expression of CSF-1 and IL-34 in their CSF-1 receptor-mediated regulation of myeloid cells. J Leukoc Biol. 2010, 88: 495-505. 10.1189/jlb.1209822.PubMedCentralCrossRefPubMed Wei S, Nandi S, Chitu V, Yeung Y-G, Yu W, Huang M: Functional overlap but differential expression of CSF-1 and IL-34 in their CSF-1 receptor-mediated regulation of myeloid cells. J Leukoc Biol. 2010, 88: 495-505. 10.1189/jlb.1209822.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Pont-Lezica L, Béchade C, Belarif-Cantaut Y, Pascual O, Bessis A: Physiological roles of microglia during development. J Neurochem. 2011, 119: 901-908. 10.1111/j.1471-4159.2011.07504.x.CrossRefPubMed Pont-Lezica L, Béchade C, Belarif-Cantaut Y, Pascual O, Bessis A: Physiological roles of microglia during development. J Neurochem. 2011, 119: 901-908. 10.1111/j.1471-4159.2011.07504.x.CrossRefPubMed
31.
Zurück zum Zitat Koushik SV, Wang J, Rogers R, Moskophidis D, Lambert NA, Creazzo TL, Conway SJ: Targeted inactivation of the sodium-calcium exchanger (ncx1) results in the lack of a heartbeat and abnormal myofibrillar organization. FASEB J. 2001, 15: 1209-1211.PubMed Koushik SV, Wang J, Rogers R, Moskophidis D, Lambert NA, Creazzo TL, Conway SJ: Targeted inactivation of the sodium-calcium exchanger (ncx1) results in the lack of a heartbeat and abnormal myofibrillar organization. FASEB J. 2001, 15: 1209-1211.PubMed
32.
Zurück zum Zitat Rigato C, Buckinx R, Le-Corronc H, Rigo JM, Legendre P: Pattern of invasion of the embryonic mouse spinal cord by microglial cells at the time of the onset of functional neuronal networks. GLIA. 2011, 59: 675-695. 10.1002/glia.21140.CrossRefPubMed Rigato C, Buckinx R, Le-Corronc H, Rigo JM, Legendre P: Pattern of invasion of the embryonic mouse spinal cord by microglial cells at the time of the onset of functional neuronal networks. GLIA. 2011, 59: 675-695. 10.1002/glia.21140.CrossRefPubMed
33.
Zurück zum Zitat Santos AM, Calvente R, Tassi M, Carrasco M-C, Martín-Oliva D, Marín-Teva JL: Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol. 2008, 506: 224-239. 10.1002/cne.21538.CrossRefPubMed Santos AM, Calvente R, Tassi M, Carrasco M-C, Martín-Oliva D, Marín-Teva JL: Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol. 2008, 506: 224-239. 10.1002/cne.21538.CrossRefPubMed
34.
Zurück zum Zitat Rymo SF, Gerhardt H, Wolfhagen Sand F, Lang R, Uv A, Betsholtz C: A two-way communication between microglial cells and angiogenic sprouts regulates angiogenesis in aortic ring cultures. PLoS One. 2011, 6: e15846-10.1371/journal.pone.0015846.PubMedCentralCrossRefPubMed Rymo SF, Gerhardt H, Wolfhagen Sand F, Lang R, Uv A, Betsholtz C: A two-way communication between microglial cells and angiogenic sprouts regulates angiogenesis in aortic ring cultures. PLoS One. 2011, 6: e15846-10.1371/journal.pone.0015846.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Kubota Y, Takubo K, Shimizu T, Ohno H, Kishi K, Shibuya M: M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis. J Exp Med. 2009, 206: 1089-1102. 10.1084/jem.20081605.PubMedCentralCrossRefPubMed Kubota Y, Takubo K, Shimizu T, Ohno H, Kishi K, Shibuya M: M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis. J Exp Med. 2009, 206: 1089-1102. 10.1084/jem.20081605.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Eilken HM, Adams RH: Dynamics of endothelial cell behavior in sprouting angiogenesis. Curr Opin Cell Biol. 2010, 22: 617-625. 10.1016/j.ceb.2010.08.010.CrossRefPubMed Eilken HM, Adams RH: Dynamics of endothelial cell behavior in sprouting angiogenesis. Curr Opin Cell Biol. 2010, 22: 617-625. 10.1016/j.ceb.2010.08.010.CrossRefPubMed
38.
Zurück zum Zitat Phng LK, Gerhardt H: Angiogenesis: A team effort coordinated by notch. Dev Cell. 2009, 16: 196-208. 10.1016/j.devcel.2009.01.015.CrossRefPubMed Phng LK, Gerhardt H: Angiogenesis: A team effort coordinated by notch. Dev Cell. 2009, 16: 196-208. 10.1016/j.devcel.2009.01.015.CrossRefPubMed
39.
Zurück zum Zitat Moya IM, Umans L, Maas E, Pereira PNG, Beets K, Francis A: Stalk cell phenotype depends on integration of notch and smad1/5 signaling cascades. Dev Cell. 2012, 22: 501-514. 10.1016/j.devcel.2012.01.007.PubMedCentralCrossRefPubMed Moya IM, Umans L, Maas E, Pereira PNG, Beets K, Francis A: Stalk cell phenotype depends on integration of notch and smad1/5 signaling cascades. Dev Cell. 2012, 22: 501-514. 10.1016/j.devcel.2012.01.007.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Larrivée B, Prahst C, Gordon E, del Toro R, Mathivet T, Duarte A: ALK1 signaling inhibits angiogenesis by cooperating with the notch pathway. Dev Cell. 2012, 22: 489-500. 10.1016/j.devcel.2012.02.005.PubMedCentralCrossRefPubMed Larrivée B, Prahst C, Gordon E, del Toro R, Mathivet T, Duarte A: ALK1 signaling inhibits angiogenesis by cooperating with the notch pathway. Dev Cell. 2012, 22: 489-500. 10.1016/j.devcel.2012.02.005.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Kim J, Oh WJ, Gaiano N, Yoshida Y, Gu C: Semaphorin 3e–plexin-d1 signaling regulates VEGF function in developmental angiogenesis via a feedback mechanism. Genes Dev. 2011, 25: 1399-1411. 10.1101/gad.2042011.PubMedCentralCrossRefPubMed Kim J, Oh WJ, Gaiano N, Yoshida Y, Gu C: Semaphorin 3e–plexin-d1 signaling regulates VEGF function in developmental angiogenesis via a feedback mechanism. Genes Dev. 2011, 25: 1399-1411. 10.1101/gad.2042011.PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Corada M, Nyqvist D, Orsenigo F, Caprini A, Giampietro C, Taketo MM: The wnt/beta-catenin pathway modulates vascular remodeling and specification by upregulating dll4/notch signaling. Dev Cell. 2010, 18: 938-949. 10.1016/j.devcel.2010.05.006.CrossRefPubMed Corada M, Nyqvist D, Orsenigo F, Caprini A, Giampietro C, Taketo MM: The wnt/beta-catenin pathway modulates vascular remodeling and specification by upregulating dll4/notch signaling. Dev Cell. 2010, 18: 938-949. 10.1016/j.devcel.2010.05.006.CrossRefPubMed
43.
Zurück zum Zitat Gore AV, Swift MR, Cha YR, Lo B, McKinney MC, Li W: Rspo1/wnt signaling promotes angiogenesis via vegfc/vegfr3. Development. 2011, 138: 4875-4886. 10.1242/dev.068460.PubMedCentralCrossRefPubMed Gore AV, Swift MR, Cha YR, Lo B, McKinney MC, Li W: Rspo1/wnt signaling promotes angiogenesis via vegfc/vegfr3. Development. 2011, 138: 4875-4886. 10.1242/dev.068460.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Shoham AB, Malkinson G, Krief S, Shwartz Y, Ely Y, Ferrara N: S1P1 inhibits sprouting angiogenesis during vascular development. Development. 2012, 139: 3859-3869. 10.1242/dev.078550.CrossRefPubMed Shoham AB, Malkinson G, Krief S, Shwartz Y, Ely Y, Ferrara N: S1P1 inhibits sprouting angiogenesis during vascular development. Development. 2012, 139: 3859-3869. 10.1242/dev.078550.CrossRefPubMed
45.
Zurück zum Zitat Jung B, Obinata H, Galvani S, Mendelson K, Ding B-S, Skoura A: Flow-regulated endothelial S1P receptor-1 signaling sustains vascular development. Dev Cell. 2012, 23: 600-610. 10.1016/j.devcel.2012.07.015.PubMedCentralCrossRefPubMed Jung B, Obinata H, Galvani S, Mendelson K, Ding B-S, Skoura A: Flow-regulated endothelial S1P receptor-1 signaling sustains vascular development. Dev Cell. 2012, 23: 600-610. 10.1016/j.devcel.2012.07.015.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Gaengel K, Niaudet C, Hagikura K, Siemsen BL, Muhl L, Hofmann JJ: The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between ve-cadherin and VEGFR2. Dev Cell. 2012, 23: 587-599. 10.1016/j.devcel.2012.08.005.CrossRefPubMed Gaengel K, Niaudet C, Hagikura K, Siemsen BL, Muhl L, Hofmann JJ: The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between ve-cadherin and VEGFR2. Dev Cell. 2012, 23: 587-599. 10.1016/j.devcel.2012.08.005.CrossRefPubMed
47.
Zurück zum Zitat Vasudevan A, Long JE, Crandall JE, Rubenstein JL, Bhide PG: Compartment-specific transcription factors orchestrate angiogenesis gradients in the embryonic brain. Nat Neurosci. 2008, 11: 429-439. 10.1038/nn2074.PubMedCentralCrossRefPubMed Vasudevan A, Long JE, Crandall JE, Rubenstein JL, Bhide PG: Compartment-specific transcription factors orchestrate angiogenesis gradients in the embryonic brain. Nat Neurosci. 2008, 11: 429-439. 10.1038/nn2074.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Simón-Marín R, Vilanova JR, Aguinagalde A, Barberá-Guillem E: Vascular architecture of the developing spinal cord in the rat: A suggested model. J Embryol Exp Morphol. 1983, 76: 27-36.PubMed Simón-Marín R, Vilanova JR, Aguinagalde A, Barberá-Guillem E: Vascular architecture of the developing spinal cord in the rat: A suggested model. J Embryol Exp Morphol. 1983, 76: 27-36.PubMed
49.
Zurück zum Zitat Greenberg DA, Jin K: From angiogenesis to neuropathology. Nature. 2005, 438: 954-959. 10.1038/nature04481.CrossRefPubMed Greenberg DA, Jin K: From angiogenesis to neuropathology. Nature. 2005, 438: 954-959. 10.1038/nature04481.CrossRefPubMed
50.
Zurück zum Zitat Fruttiger M: Development of the retinal vasculature. Angiogenesis. 2007, 10: 77-88. 10.1007/s10456-007-9065-1.CrossRefPubMed Fruttiger M: Development of the retinal vasculature. Angiogenesis. 2007, 10: 77-88. 10.1007/s10456-007-9065-1.CrossRefPubMed
51.
Zurück zum Zitat Checchin D, Sennlaub F, Levavasseur E, Leduc M, Chemtob S: Potential role of microglia in retinal blood vessel formation. Invest Ophthalmol Vis Sci. 2006, 47: 3595-3602. 10.1167/iovs.05-1522.CrossRefPubMed Checchin D, Sennlaub F, Levavasseur E, Leduc M, Chemtob S: Potential role of microglia in retinal blood vessel formation. Invest Ophthalmol Vis Sci. 2006, 47: 3595-3602. 10.1167/iovs.05-1522.CrossRefPubMed
52.
Zurück zum Zitat Unoki N, Murakami T, Nishijima K, Ogino K, van Rooijen N, Yoshimura N: SDF-1/CXCR4 contributes to the activation of tip cells and microglia in retinal angiogenesis. Invest Ophthalmol Vis Sci. 2010, 51: 3362-3371. 10.1167/iovs.09-4978.CrossRefPubMed Unoki N, Murakami T, Nishijima K, Ogino K, van Rooijen N, Yoshimura N: SDF-1/CXCR4 contributes to the activation of tip cells and microglia in retinal angiogenesis. Invest Ophthalmol Vis Sci. 2010, 51: 3362-3371. 10.1167/iovs.09-4978.CrossRefPubMed
53.
Zurück zum Zitat Stefater JA, Lewkowich I, Rao S, Mariggi G, Carpenter AC, Burr AR: Regulation of angiogenesis by a non-canonical wnt-flt1 pathway in myeloid cells. Nature. 2011, 474: 511-515. 10.1038/nature10085.PubMedCentralCrossRefPubMed Stefater JA, Lewkowich I, Rao S, Mariggi G, Carpenter AC, Burr AR: Regulation of angiogenesis by a non-canonical wnt-flt1 pathway in myeloid cells. Nature. 2011, 474: 511-515. 10.1038/nature10085.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O: Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature. 2012, 484: 110-114. 10.1038/nature10908.CrossRefPubMed Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O: Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature. 2012, 484: 110-114. 10.1038/nature10908.CrossRefPubMed
55.
Zurück zum Zitat Tammela T, Zarkada G, Wallgard E, Murtomäki A, Suchting S, Wirzenius M: Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature. 2008, 454: 656-660. 10.1038/nature07083.CrossRefPubMed Tammela T, Zarkada G, Wallgard E, Murtomäki A, Suchting S, Wirzenius M: Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature. 2008, 454: 656-660. 10.1038/nature07083.CrossRefPubMed
56.
Zurück zum Zitat Tammela T, Zarkada G, Nurmi H, Jakobsson L, Heinolainen K, Tvorogov D: VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing notch signalling. Nat Cell Biol. 2011, 13: 1202-1213. 10.1038/ncb2331.PubMedCentralCrossRefPubMed Tammela T, Zarkada G, Nurmi H, Jakobsson L, Heinolainen K, Tvorogov D: VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing notch signalling. Nat Cell Biol. 2011, 13: 1202-1213. 10.1038/ncb2331.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Outtz HH, Tattersall IW, Kofler NM, Steinbach N, Kitajewski J: Notch1 controls macrophage recruitment and notch signaling is activated at sites of endothelial cell anastomosis during retinal angiogenesis in mice. Blood. 2011, 118: 3436-3439. 10.1182/blood-2010-12-327015.PubMedCentralCrossRefPubMed Outtz HH, Tattersall IW, Kofler NM, Steinbach N, Kitajewski J: Notch1 controls macrophage recruitment and notch signaling is activated at sites of endothelial cell anastomosis during retinal angiogenesis in mice. Blood. 2011, 118: 3436-3439. 10.1182/blood-2010-12-327015.PubMedCentralCrossRefPubMed
58.
Zurück zum Zitat Hofmann JJ, Luisa I-AM: Notch expression patterns in the retina: An eye on receptor-ligand distribution during angiogenesis. Gene Expr Patterns. 2007, 7: 461-470. 10.1016/j.modgep.2006.11.002.PubMedCentralCrossRefPubMed Hofmann JJ, Luisa I-AM: Notch expression patterns in the retina: An eye on receptor-ligand distribution during angiogenesis. Gene Expr Patterns. 2007, 7: 461-470. 10.1016/j.modgep.2006.11.002.PubMedCentralCrossRefPubMed
Metadaten
Titel
The importance of microglia in the development of the vasculature in the central nervous system
verfasst von
Tom Arnold
Christer Betsholtz
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Vascular Cell / Ausgabe 1/2013
Elektronische ISSN: 2045-824X
DOI
https://doi.org/10.1186/2045-824X-5-4

Weitere Artikel der Ausgabe 1/2013

Vascular Cell 1/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.