You are on page 1of 32

ANNUAL REVIEWS

Further

Click here for quick links to Annual Reviews content online, including: Other articles in this volume Top cited articles Top downloaded articles Our comprehensive search

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Connecting Vascular and Nervous System Development: Angiogenesis and the Blood-Brain Barrier
Stephen J. Tam and Ryan J. Watts
Neurodegeneration Labs, Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080; email: watts.ryan@gene.com

Annu. Rev. Neurosci. 2010. 33:379408 First published online as a Review in Advance on April 1, 2010 The Annual Review of Neuroscience is online at neuro.annualreviews.org This articles doi: 10.1146/annurev-neuro-060909-152829 Copyright c 2010 by Annual Reviews. All rights reserved 0147-006X/10/0721-0379$20.00

Key Words
endothelial cells, axon guidance, neurovascular unit, astrocytes, tight junctions, Wnt signaling

Abstract
The vascular and nervous systems share a common necessity of circuit formation to coordinate nutrient and information transfer, respectively. Shared developmental principles have evolved to orchestrate the formation of both the vascular and the nervous systems. This evolution is highlighted by the identication of specic guidance cues that direct both systems to their target tissues. In addition to sharing cellular and molecular signaling events during development, the vascular and nervous systems also form an intricate interface within the central nervous system called the neurovascular unit. Understanding how the neurovascular unit develops and functions, and more specically how the bloodbrain barrier within this unit is established, is of utmost importance. We explore the history, recent discoveries, and unanswered questions surrounding the relationship between the vascular and nervous systems with a focus on developmental signaling cues that guide network formation and establish the interface between these two systems.

379

Contents
INTRODUCTION . . . . . . . . . . . . . . . . . . ANATOMICAL PATTERN OF THE VASCULAR AND NERVOUS SYSTEMS . . . . . . . . . . . . Evolution of the Nervous and Vascular Systems . . . . . . . . . . . Shared Anatomical Patterning . . . . . . SHARED CELLULAR MECHANISMS . . . . . . . . . . . . . . . . . . . The Cellular Organization of the Nervous and Vascular Systems. . . The Axon Growth Cone and Endothelial Tip Cell . . . . . . . . . . . . SHARED MOLECULAR MECHANISMS . . . . . . . . . . . . . . . . . . . Axon Guidance Molecules and Angiogenesis . . . . . . . . . . . . . . . The Slits and Robos in Vascular Guidance . . . . . . . . . . . The Semaphorins/Plexins/Nrps in Vascular Guidance . . . . . . . . . . . The Netrin/Unc5/DCC Family in Vascular Guidance . . . . . . . . . . . The Ephrin/Eph Family in Vascular Guidance . . . . . . . . . . . THE INTERFACE BETWEEN THE VASCULAR AND NERVOUS SYSTEMS . . . . . . . . . . . . . . . . . . . . . . . . Molecular Properties of the Blood-Brain Barrier . . . . . . . . . . . . . The Neurovascular Unit . . . . . . . . . . . CNS Angiogenesis and BBB Formation: A Role for Wnt Signaling . . . . . . . . . . . . . . . . . . . . . . . CONCLUSIONS AND FUTURE DIRECTIONS . . . . . . . . . . . . . . . . . . . . 380

381 381 382 384 384 385 386 386 388 388 390 391

392 392 395

397 401

CNS: central nervous system NVU: neurovascular unit BBB: blood-brain barrier

INTRODUCTION
The anatomical similarities between the vascular and nervous systems have been recognized for centuries. Recent discoveries have continued to deepen this apparent relationship at both the cellular and the molecular
Tam

levels. At a gross anatomical level, the pathways of blood vessels and nerves appear congruous. At the cellular level, extending vessels and neurons utilize similar specialized structures that drive target tissue guidance through sensing their local environment. Ultimately these similarities can be traced back to several families of shared molecular guidance cues that guide both endothelial cells and neurons (Carmeliet & Tessier-Lavigne 2005, Dickson 2002, Larriv ee et al. 2009, ODonnell et al. 2009, Tessier-Lavigne & Goodman 1996). Although still in its early years, the eld of vascular guidance has taken many cues from past insights provided by the axonal guidance principles of attraction and repulsion. Beyond the common anatomical, cellular, and molecular properties, the vascular and nervous systems have developed an intricate relationship within the central nervous system (CNS) itself. This connection is found at the interface between the vascular and nervous systems, generally known as the neurovascular unit (NVU). The NVU is composed of endothelial cells, pericytes, glia, and neurons, which are tightly coupled to control cerebrovascular function (Hawkins & Davis 2005, Iadecola 2004). A major component of this interface is the blood-brain barrier (BBB), which is formed by endothelial cells of the NVU. The specialized blood vessels of the BBB are characterized by the formation of tight junctions, expression of specialized transporters and pumps, and reduced basal endocytic activity as compared with vessels found in peripheral organs (Rubin & Staddon 1999). Although the barrier is generally termed the blood-brain barrier, there are in fact several distinct barriers between the periphery and the nervous system, including the blood-cerebral spinal uid barrier, the bloodretinal barrier, the bloodspinal cord barrier, and other barriers. Furthermore, this barrier is, in fact, a dynamic site of molecular transport that constantly regulates the ow of essential components, such as amino acids, across endothelial walls. Although many fundamental questions regarding BBB development and maintenance remain unanswered, the crucial

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

380

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

role of the BBB in the protection and proper functioning of the nervous system has begun to attract the attention of researchers. Recent discoveries have uncovered several molecular cues that regulate barrier formation. For instance, the wingless-type protein (Wnt) signaling pathway was identied as a key activator of angiogenesis in the nervous system and subsequent BBB formation (Daneman et al. 2009, Liebner et al. 2008, Stenman et al. 2008). Notably, Wnt signaling has been extensively studied in nervous system development and function (Salinas & Zou 2008). The identication of Wnt signaling as a regulator of angiogenesis and BBB formation in the CNS provides molecular evidence to support the original observation that neuronal tissue provides instructive cues to advancing endothelial cells, forming specialized blood vessels in the nervous system (Stewart & Wiley 1981). These ndings are also another example of the common signaling pathways that drive the development of both the vascular and the nervous systems. Signicant progress has been made in identifying how vascular and nervous systems develop and function; recent advances highlight the common cellular and molecular mechanisms shared by these two systems. We examine these similarities and highlight the close relationship between the vascular and nervous systems, with an emphasis on their shared common developmental principles and how the interface between these systems, the BBB, is formed.

ANATOMICAL PATTERN OF THE VASCULAR AND NERVOUS SYSTEMS Evolution of the Nervous and Vascular Systems
The fundamental requirement for an organism to both sense its environment and broadly distribute nutrients throughout its body has driven the evolution of the nervous and vascular systems, respectively. Although the exact ori-

gin of the nervous system is debatable, investigators generally believe that modern nervous systems arose rst in the form of diffuse nerve nets, in sea-dwelling organisms such as cnidarians (Miller 2009). These organisms lack a vascular system (dened as a system to carry both oxygen and nutrients) because resources were readily available through diffusion and ingestion. On the basis of these observations, the general consensus indicates that the modern nervous system evolved prior to the vascular system. The major driving force behind vascular evolution is the need for oxygen to fuel aerobic metabolism in multicellular organisms, combined with the limited ability of oxygen to diffuse through tissue (Fisher & Burggren 2007). For arthropods, two systems seem to have evolved independently to distribute nutrients and oxygen. To deliver nutrients, hemolymph bathes all cells and is distributed by the coordinated movement of muscles and a primitive heart. Independent of nutrients, oxygen is delivered by the tracheal system. The tracheal system of insects is most akin to the modern vascular system as it relates to developmental principles, highlighted by the stereotyped formation of a tubular network that functions by directing air exchange of oxygen and carbon dioxide. In fact, many features of tube formation and sprouting are shared between the insect tracheal system and the modern vascular systems (Lubarsky & Krasnow 2003). As larger organisms evolved, however, a system to distribute oxygen and nutrients throughout dense tissue more efciently became necessary. Consistent with the chronology of neuronal and vascular evolution, several vascular-specic genes may have arisen later in evolution. Vascular endothelial growth factor (VEGF) was rst discovered as a growth factor that selectively mediates endothelial proliferation and vessel permeability (Keck et al. 1989, Leung et al. 1989), and expression of this hypoxiaresponsive gene is tightly regulated to control vasculogenesis and angiogenesis (Coultas et al. 2005, Fong 2009). Distant homologs to both mammalian VEGF and VEGFRs do exist

VEGF: vascular endothelial growth factor

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

381

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

in invertebrates (Cho et al. 2002). The functions of these molecules in Drosophila are limited to controlling blood cell migration, a function that is speculated to be a precursor to the many VEGF-driven functions in mammals. One of the coreceptors for VEGF, neuropilin-1 (Nrp1), arose after the nervous system in evolution. Nrp1 was rst identied as a receptor for the axon guidance cue semaphorin (He & Tessier-Lavigne 1997, Kolodkin et al. 1997) and then subsequently shown also to be a receptor for VEGF (Soker et al. 1998). Nrp1 is found in vertebrates but is absent from invertebrates, whereas semaphorins and their other coreceptors, the plexins, are present in both invertebrates and vertebrates (Kolodkin 1996, Kolodkin et al. 1992, Tamagnone et al. 1999). Therefore, the evolution of the Nrp1 gene may have been congruent with the origin of the modern vascular system, yet Nrp1 is an essential component in vertebrate neuronal development as well. Nrp1 interaction with both semaphorin and VEGF is one example of how the vascular and nervous systems have co-opted the same molecular cascades for development. However, many of the guidance cues may have originated rst in the nervous system, with family members likely evolving later to play a role in vascular development. These pathways are discussed in more detail in subsequent sections.

Shared Anatomical Patterning


Similar patterning between the vascular and nervous systems is apparent at the anatomical level (Figure 1a). These similarities were likely rst observed at the macroscopic level (Vesalius

1543). With modern histological methodology, the coursing of artery and veins with nerve bundles can now be described at the microscopic level (Larriv ee et al. 2009, Mukouyama et al. 2002). How can such highly stereotypic patterning in two diverse systems be achieved? Do developing vessels provide signals for growing axons, or vice versa? Looking at the developing mouse embryo, one can see that vascular outgrowth precedes axon outgrowth (Figure 1b). Consistent with this observation, many molecular cues are provided by the developing vasculature to guide growing axons. For example, endothelin-3 is secreted by smooth-muscle cells of the external carotid artery, providing an attractive signal for extending axons of the superior cervical ganglia (Makita et al. 2008). Furthermore, vascular smooth-muscle expression of Artemin, a glial cellderived neurotrophic factor (GDNF) family member, is a signal attracting axons of sympathetic neurons (Honma et al. 2002). Artemin-decient mice show axon outgrowth and patterning defects, an observation that is phenocopied in mice lacking the Artemin receptor GFRalpha3. These ndings provide convincing evidence that vessels can direct neurons to form congruent patterns. As part of normal vascular and neural development, exuberant primitive networks are initially formed followed by the selective remodeling of these networks to form functional circuits. In the case of vascular remodeling, a robust vascular plexus is generated in the embryo, followed by remodeling to arteries, veins, and ne capillary networks. Vascular remodeling takes place after peripheral nerve development and is directed by peripheral sensory neurons and Schwann cells (Mukouyama et al. 2002).


Figure 1 Anatomical resemblance between the vascular and nervous systems. (a) Depiction of the shared anatomical patterning between nervous and vascular networks in the human body. Recent evidence indicates that blood vessels and neurons direct their outgrowth using shared molecular mechanisms, ultimately resulting in a similarly patterned network. (b) Although developing mouse embryos exhibit shared nervous and vascular patterning as well, the vascular system develops at a faster pace than does the nervous system. In fact, vessels can direct neurons to form congruent patterns. Left panel: neurons visualized by neurolament (Nf) staining of an E12.5 embryo. Right panel: blood vessels visualized by FLK1 in situ hybridization staining of an E10.5 embryo.
382 Tam

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Nf Nervous system Circulatory system

FLK1

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

383

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

These conclusions are derived from both necessity and sufciency experiments. Mutant embryos that lack peripheral neurons do not undergo proper vascular remodeling, highlighted by defects in arterial differentiation. Moreover, mutants that misdirect peripheral axon outgrowth result in coalignment of blood vessels with these misguided neuronal patterns. Mukouyama et al. (2005) later showed that VEGF is one such cue involved in vascular remodeling and arteriogenesis that is provided by neurons and Schwann cells. More than four and a half centuries since Vesalius rst observed the overall anatomical similarities between vascular and neuronal networks, evidence now demonstrates that vessels and neurons codirect their patterning to achieve exquisite alignment at the anatomical level. For such guidance to take place, growing vessels and neurons must be able to sense the complex chemical cues that orchestrate the morphological changes necessary for proper patterning. These sensory mechanisms are found in the cellular structures that constitute the developing neuron and blood vessel.

SHARED CELLULAR MECHANISMS The Cellular Organization of the Nervous and Vascular Systems
In 1906, Camillo Golgi and Santiago Ramon y Cajal presented differing theories about the cellular organization of the nervous system in a famous Nobel debate. Golgi postulated that the nervous system consisted of a syncytial system with multiple cells of the nervous system possibly connecting via protoplasmic fusions to form a reticular system (Golgi 1906). Cajal, on the other hand, was an ardent proponent of the neuron doctrine, which argued that cells in the nervous system constitute individual components connected by a granular cement, or special conducting substance keeping neurons intimately in contact, yet in contiguity but not in continuity (Ramon y Cajal 1906). History has largely proven both Cajal
384 Tam

and Waldeyer, the rst to propose the neuron doctrine, to be correct. We understand now that the nervous system consists of a complex network of diverse neurons, with extending axons connecting to dendrites of target cells via specialized contacts known as synapses. Many neurons must send axonal processes long distances through a labyrinth of tissue to reach their nal targets. The emergence of embryonic neurons from neuroblasts, followed by their migration and elaboration, results in single cellular units that possess the ability to receive and send information. The nervous system also contains a battery of glial cells, which are required for a multitude of nervous system functions, including enhancement of nerve conductance, immune surveillance, and neurotransmitter processing and signaling (Barres 2008). Golgi himself rst observed that glial cells make direct contacts with the vasculature, which were likely to be the astrocytic endfeet and their ensheathment of blood vessels (see The Neurovascular Unit, below). Oligodendrocytes and Schwann cells are responsible primarily for myelination of the central and peripheral nervous systems, respectively. They are functionally similar to pericytes and smooth-muscle cells of the vasculature because both are supporting cell types that enhance the function of the main constituent cell types, neurons and endothelial cells, respectively (Bergers & Song 2005, Betsholtz et al. 2005). However, it remains to be determined if similar signaling mechanisms are employed toward the formation of both endothelialpericytes and neuron-glia interactions. In contrast to network formation in the nervous system, which is the result of elaborated single neurons extending protoplasmic processes to form specic connections, the cellular components of the vascular system undergo blood vessel formation through two distinct processes. First, in vasculogenesis, endothelial cells differentiate from angioblasts to form a vascular plexus, which consists of a meshwork of connected strands of endothelial cells (Coultas et al. 2005). The vascular plexus serves as the starting substrate for further growth and

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

renement. Second, new vessels sprout from existing vessels, a process termed angiogenesis. VEGF is necessary for both vasculogenesis and angiogenesis; however, other molecules provide additional cues for further differentiation and renement of the vascular system. Similar to the nervous system, where exuberant connectivity is followed by pruning to form a functional network, the vascular system also undergoes remodeling. This process is inuenced by both blood ow and contact with surrounding tissues. Furthermore, vessels formed via angiogenesis must sense their environment to follow the correct trajectory toward their nal target.

The Axon Growth Cone and Endothelial Tip Cell


Cajals illustrious career was also highlighted by his discovery of the axon growth cone, a

structure found at the tip of growing axons that guides outgrowth by sensing its local environment (de Castro et al. 2007). In Cajals own words, a growth cone is like a living batteringram, soft and exible, which advances mechanically, pushing aside the obstacles that it nds in its way until it reaches its peripheral destination (Ramon y Cajal 1917). Modern imaging has since veried Cajals assessment and has further dened the molecular components of the growth cone (Lowery & Van Vactor 2009). Specically, an axonal growth cone is found at the end of an axon shaft and consists largely of cytoskeletal machinery (Figure 2a). The hand-like structure itself is composed of both actin-rich lopodia and lamellopodia, with actin bundles that are connected to the microtubule framework that continues down the axon shaft. More than 100 years after Cajals original description of the axon growth cone, Gerhardt

Axon growth cone

Endothelial tip cell


Attractive guidance cues

a
Filopodium

Attractive guidance cues

Filopodium Lamellipodium Endothelial stalk cells

LamellIpodium

on Ax
Repulsive guidance cues
Figure 2

Capillary lumen (blood)

Endothelial tip cell Repulsive guidance cues

The axon growth cone and endothelial tip cell. Blood vessels and neurons must sense their environment to follow the correct trajectory toward their nal target. Cellular projections of axons and endothelia that guide such outgrowth share morphological features. (a) The axon growth cone forms at the leading edge of migrating axons through an elaborate cytoskeletal actin network linked to axonal microtubules. (b) The endothelial tip cell caps the migrating end of stalk cells, a lumenized column of cells that form a budding or angiogenic blood vessel. Although the tip cell is its own cellular entity separate from the rest of the growing blood vessel, it is the functional analog of the axonal growth cone. Both lopodial and lamellopodial projections of the extending growth cone and tip cell are believed to form the motor that physically pulls cells toward attractive cues ( green) and away from repulsive cues (red) as extending neurons and blood vessels sense their environment.
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 385

et al. (2003) described a similar structure in angiogenic sprouts of growing blood vessels in the mouse retina. This structure is composed of a specialized cell found at the growing end of extending vessels, termed the endothelial tip cell (Figure 2b). The shaft of the extending vessel is composed of a lumenized endothelial cell chain made of stalk cells. From an architectural standpoint, the tip cell and associated endothelial stalk cells are similar to the axon growth cone and its associated axonal shaft. Tip cells, akin to axon growth cones, are actin-rich structures that dynamically navigate their environment when sprouting from existing vasculature. This process was elegantly observed through live imaging of green uorescent protein (GFP)-expressing vessels in transgenic zebrash, where extending tip cells were described as functionally similar to axon growth cones (Lawson & Weinstein 2002). Tip cells, at least in the retina, are exquisitely responsive to VEGF (Gerhardt et al. 2003). Using mice engineered to express distinct isoforms of VEGF that have different diffusion properties, investigators showed that endothelial tip cells migrate in response to VEGF gradients, whereas stalk cell proliferation is a property of total VEGF concentrations. These data are consistent with previous observations utilizing VEGF isoform mutants (Ruhrberg et al. 2002). Specically, a shallow VEGF gradient with high total concentrations of VEGF exposure results in large-diameter vessels with reduced branching. VEGF receptor 2 (Flk1) is both expressed on the tip cell and necessary for lopodia formation (Gerhardt et al. 2003). Thus, in the retina, VEGF acts as the predominant mitogenic factor and is chemoattractive for blood vessels. How is a tip cell selected to form an angiogenic sprout? The vascular system likely addressed this need by co-opting the Notch signaling cascade described rst in Drosophila. In the case of y tracheal development, a leading tracheal cell is selected by Notch-mediated lateral inhibition (Ghabrial & Krasnow 2006). In mammals, delta-like 4 (Dll4) appears to be the major Notch ligand regulating vascular
386 Tam

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

development (Duarte et al. 2004, Gale et al. 2004, Krebs et al. 2004). Additional mechanistic studies have carefully dissected Dll4s role in vascular development and concluded that Dll4/Notch signaling is likely acting via lateral inhibition to establish the tip cell morphology, whereas adjacent cells are destined to become stalk cells (Hellstrom et al. 2007, Lobov et al. 2007, Suchting et al. 2007). These observations should not be surprising because Notch signaling is a major mechanism for cell fate determination in many tissues, including the nervous system (Gaiano & Fishell 2002). The cellular parallels between an extending neurons axon growth cone and an angiogenic blood vessels tip cell suggest that these structures may respond in ways similar to chemical cues provided by their respective environments. In 1963, Roger Sperry summarized the chemospecicity hypothesis, which postulates that chemical cues in the target tissue provide specic directions for the developing and regenerating nervous system, resulting in stereotyped neural network formation (Sperry 1963). Do blood vessels also respond to chemical cues resulting in stereotyped development? Recent discoveries have addressed this hypothesis at the molecular level and provide strong evidence that the nervous and vascular systems share common principles of development.

SHARED MOLECULAR MECHANISMS Axon Guidance Molecules and Angiogenesis


The possibility that the vascular and nervous systems respond to common molecular cues to guide their development is highly plausible owing to both their stereotyped circuitry and the shared common cellular structures used to sense their environment during development. We have described evidence that nerves may provide cues to growing vessels; likewise, the opposing relationship has also been established. A major theme of cellular guidance is the balance between attractive and repulsive forces

Watts

(Figure 2). In introducing the endothelial tip cell, we have discussed how VEGF is a major attractive molecule for extending blood vessels (Gerhardt et al. 2003). Do other attractive cues exist for blood vessels? Are there repulsive forces that also guide developing vessels? In searching for additional candidate molecules, we now turn our attention to the accumulating evidence indicating that the classical axon guidance cues also guide vascular tip cells. Axon guidance molecules exist in four major families: (a) Slit/Robo, (b) semaphorin/ plexin/neuropilin, (c) Netrin/Unc5/DCC, and
Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

(d) Ephrin/Eph (Figure 3). The role of these pathways in guidance of axons in the developing nervous system has been described extensively (Dickson 2002, ODonnell et al. 2009, Tessier-Lavigne & Goodman 1996). Recent studies also indicate that various well-described morphogens, including wingless-type proteins (Wnts), bone morphogenic proteins (BMPs), and sonic hedgehog (Shh) also mediate axon guidance (Butler & Dodd 2003, Charron et al. 2003, Lyuksyutova et al. 2003, Yoshikawa et al. 2003). With the exception of the Wnts, which are discussed in detail below related to

Vascular guidance molecules


EphrinB2 Other Ephrins Cell membrane Slit1-3 Sema3A, 3C, 3F
Class 4 and 5 Semas

VEGFs
Netrin1-4

Robo4
Robo1-3

Neuropilins
DCC

EphB4 PlexinD1
Other Plexins

VEGFRs

Unc5b

Other Ephs

Figure 3 Axon guidance pathways involved in angiogenesis. Schematic representation of the four major families of axon guidance ligand-receptor pairs: Slit/Robo, semaphorin/plexin/neuropilin, Netrin/Unc5/DCC, and Ephrin/Eph. Axonal guidance cues mediate complex cellular navigational programs within axons, as both chemoattractants and repellents. Recent evidence has identied roles for several of these classic guidance molecules in directing angiogenic tip cells toward their nal destination.
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 387

angiogenesis and BBB formation in the developing CNS, we discuss the role of the four major families of axon guidance molecules in angiogenesis.

The Slits and Robos in Vascular Guidance


The Slit/Robo family of axon guidance molecules was discovered as mediators of commissural axon crossing in Drosophila (Kidd et al. 1998, 1999; Seeger et al. 1993) and of axon guidance in C. elegans (Zallen et al. 1998). Wang et al. (1999) showed that Slit enhances axonal branching and elongation (Wang et al. 1999). There are three members of the Slit ligand family and four members of the Robo receptor family (Figure 3). Slits contain 4 leucine-rich repeats, 79 EGF repeats, and one laminin domain. The roundabout (Robo) receptors were named after their y loss-of-function phenotype of aberrant midline crossing, including axons repeatedly crossing the midline, giving the appearance of a circular road junction, or roundabout, when analyzed by histology (Seeger et al. 1993). The Robo1-3 receptors consist of ve Ig domains, three bronectin type 3 domains, and one intracellular signaling domain. A fourth member of the Robo receptor family, Robo4 or magic roundabout, contains only two Ig domains and two bronectin type 3 domains. Notably, Robo4 is almost exclusively expressed in the vasculature (Huminiecki et al. 2002). During neural development, Slit/Robo interactions can result in either a repulsive or an attractive response (Dickson & Gilestro 2006). Most data indicate that Slit/Robo binding on neurites primarily regulates a repulsive signal. Thus it was intriguing when Wang et al. (2003) found Slit2 to be an attractive factor for extending angiogenic sprouts through binding to Robo1 on endothelial cells. In contrast to this data, others have argued that Slit2 is repulsive to endothelial cells via binding to Robo4 (Park et al. 2003). To complicate matters further, it is unclear whether Slit2 binds to Robo4 because in vitro binding studies showed a convincing
388 Tam

interaction only between Slit2 and Robo1, but not to Robo4 (Suchting et al. 2005). Robo4 may also mediate a repulsive or attractive response on the basis of cellular context, including cell-type and/or ligand binding. For example, Sheldon et al. (2009) recently showed that Robo1 and Robo4 can form heterodimers, indicating that Slit2 binding to Robo1 may result in signaling via Robo4. In addition to in vitro observations for Slit/Robo interactions in the context of the vascular system, studies in zebrash have shown that a knockdown of Robo4 results in an intersomitic vessel guidance defect with a reduction in vessel sprouting and a misdirection of those sprouts that do form (Bedell et al. 2005). However, no example of a vascular developmental defect in mouse knockouts of the various Slit/Robo genes has been reported. A recent study has proposed that Robo4 mediates vascular integrity in adult animals by inhibiting angiogenesis and reducing vascular permeability (Jones et al. 2008). Robo4-decient mice exhibited an increase in vascular sprouting and permeability in an oxygen-induced retinopathy model. Furthermore, injecting Slit2 reversed the increase in angiogenesis and vascular permeability in these models in a Robo4dependent fashion. These data support a repulsive role for Robo4 in angiogenic sprouting. However, because Robo4 is expressed on endothelial stalk cells in the retina, it is intriguing to speculate that Robo4 may inhibit new tip cell formation from existing vessels. To date, the interaction between Robo4 and Slit2 remains to be fully veried, leaving open the possibility that other ligands mediate Robo4 function.

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

The Semaphorins/Plexins/Nrps in Vascular Guidance


Although semaphorins (Sema) were rst discovered as mediators of axon growth cone collapse (Raper 2000), they are now more commonly recognized as proteins with robust effects on cellular morphology. There are eight classes of Sema proteins, including both membrane-bound and secreted forms. For the

Watts

purposes of this review, we focus on one of the most versatile families, the class 3 semaphorins (Sema3). The Sema3 family members are secreted proteins that contain a Sema domain, a plexin/semaphorin/integrin domain (PSI), and an Ig domain (Figure 3). The Plexin and Neuropilin receptor families collaborate to form multimeric receptor assemblies responsible for downstream Sema signaling. The extracellular region of plexins consists of one Sema domain, three PSI domains, four Ig-like/plexins/transcription factor domains (IPT), and two Ig domains. A transmembrane helix links this region to the cytoplasmic region containing one coiled-coil helix and one Racbinding domain (RBD) that bisects a GTPase activating protein domain (GAP). Mammals have two neuropilin (Nrp) proteins: Nrp1 and Nrp2. Nrps share two complement-binding domains (CUB or a1a2 domains), two coagulation factor V/VIII homology domains (b1b2), and a meprin/A5 protein/phosphatase- domain (MAM) in the extracellular space followed by a transmembrane helix and a short cytoplasmic tail. Furthermore, Nrps can also be expressed as soluble isoforms lacking the MAM and transmembrane domains (Rossignol et al. 2000). In addition to being semaphorin receptors, Nrps bind multiple VEGF isoforms and mediate distinct VEGF functions (Pan et al. 2007; Soker et al. 1998, 2002). The Sema/Plexin/Nrp family is a prime example of a group of molecules with diverse functions in both neuronal and vascular development. Here, we focus on their roles in vascular biology. Sema3s were originally classied as potent axonal chemorepellents that signal indirectly through Plexin receptors by binding to Nrps (Chen et al. 1997, Fujisawa 2004, He & TessierLavigne 1997, Kolodkin et al. 1997). Of the seven Sema3s (A-G) reported in mammals, several function in more versatile ways, including a key role in guiding extending endothelial cells (Tran et al. 2007). Sema3A was rst described in the vascular system as a mediator of vessel tip cell lamellipodial collapse and was thus an

inhibitor of angiogenesis (Miao et al. 1999, Serini et al. 2003, Shoji et al. 2003). However, a role for Sema3A in endothelial biology and vascular development has been disputed. More recent studies have shown that Sema3A does not reduce endothelial cell migration (Pan et al. 2007), nor does it compete with VEGF for binding to Nrp1 (Appleton et al. 2007). Furthermore, sema3a/ knockout mice do not have vascular development phenotypes (Vieira et al. 2007). Additional studies may be needed to determine if Sema3A plays a more specic or subtle role in vascular biology that has yet to be discovered. For example, although the overwhelming evidence suggests that semaphorins act as chemorepulsive cues in the nervous system, at least one example shows that the cellular context can switch Sema from a repulsive to an attractive signal (Song et al. 1998). In contrast to Sema3A, Sema3E is emerging as a critical mediator of vascular biology through its direct interaction with the endothelial expressed receptor, PlexinD1 (Gitler et al. 2004, Gu et al. 2005, Torres-Vazquez et al. 2004). The vascular expression of plexinD1 was rst described in two separate studies, one examining all Plexins and focusing on their roles in the nervous system (Cheng et al. 2001) and the other looking specically at plexinD1 expression during embryonic development (van der Zwaag et al. 2002). Sema3e and plexinD1 loss-of-function studies in both zebrash and mouse result in a vessel guidance defect consistent with a repulsive role for Sema3E acting via PlexinD1. Somites express Sema3E, thereby preventing the PlexinD1expressing intersomitic vessels from entering the somites. However, plexinD1 knockout mice show a cardiac development defect not observed in sema3E knockouts, thus suggesting that another Sema, speculated to be Sema3C, may act through PlexinD1. Nrp receptors bind both VEGF and class 3 semaphorins. It is therefore not surprising that knockouts of both Nrp1 and Nrp2, which are expressed in developing vasculature and lymphatic systems respectively, lead to numerous vessel-patterning and lymphangiogenic
389

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

defects (Gu et al. 2003, Kawasaki et al. 1999, Takashima et al. 2002). However, Nrps interactions with both VEGFs and Sema3s complicate the interpretation of these results, making it difcult to pinpoint a particular phenotype as being related to Sema or VEGF. To address this issue Gu and colleagues (2002) rst performed structure/function studies to identify residues responsible for Sema3 binding to Nrp1. Subsequently, knock-in mice expressing mutant Nrp1 lacking Sema binding and keeping VEGF binding intact were generated (Gu et al. 2003). These mice showed no vascular defects, suggesting that Nrp1s interaction with VEGF, but not Sema, is critical for vascular patterning. On the basis of Nrp1s multifaceted role in vascular biology, this molecule is now being therapeutically targeted to modulate angiogenesis in vivo.

The Netrin/Unc5/DCC Family in Vascular Guidance


Netrin was rst identied as UNC-6 in C. elegans (Ishii et al. 1992), and three Netrin family members have been identied in mammals (Moore et al. 2007). The netrins are secreted proteins that contain one laminin domain, three EGF-like repeats, and a carboxylterminal domain (CRD) (Figure 3). Netrins act as both chemoattractants and chemorepellents using their interactions with respective receptors. During CNS development, Netrin1 is secreted from cells at the ventral midline to attract commissural axons (Kennedy et al. 1994, Serani et al. 1994) via activation of the DCC (Deleted in Colorectal Cancer) receptor (Keino-Masu et al. 1996). In netrin1 and dcc knockout mice, most commissural axons fail to reach and cross the midline (Fazeli et al. 1997, Serani et al. 1996). DCC and neogenin are members of the DCC family of receptors and contain four Ig domains, six Fn3 domains, and one transmembrane domain, followed by an intracellular signaling domain with putative protein interaction and phosphorylation sites (Moore et al. 2007). DCC can also interact with Robo
390 Tam

receptors to inactivate Netrins attractant activity once midline crossing has been achieved (Stein & Tessier-Lavigne 2001). Conversely, Netrin-mediated axon repulsion involves the activation of Unc5 (uncoordinated 5) receptor family members, either as homodimers or Unc5-DCC heterodimers (Hong et al. 1999, Keleman & Dickson 2001, Leonardo et al. 1997). The Unc5 family contains two Ig domains, two thrombospondin type I domains, one transmembrane domain, one Zona occludens-5 domain, one DCC interacting domain, and one death domain. Netrins and their cognate receptors have also been implicated in the regulation of several developmental processes in nonneuronal tissues. The rst hints that Netrin/Unc5 signaling may regulate blood vessel morphogenesis were discovered as a consequence of knocking out the Unc5b receptor (Lu et al. 2004). Depletion of Unc5b in mice and zebrash resulted in excessive tip cell lopodia formation and branching, suggesting that Unc5b activation drives endothelial repulsion. Investigators showed that Unc5b is present in the tip cell of extending blood vessels (Bouvr ee et al. 2008, Larriv ee et al. 2007). Knockdown of netrin1a in zebrash results in a loss-of-function phenotype similar to the unc5b loss-of-function. Similar to Sema3A and PlexinD1, intersomitic paths express Netrin1a and thereby prevent the Unc5b-expressing tip cells from meandering off path into the somites. In mice, the ligand for Unc5b-mediated vessel tip cell repulsion remains to be identied because netrin1 knockouts maintain normal vascular architecture (Salminen et al. 2000, Serani et al. 1996). The lack of Netrin1 involvement in mammalian blood vessel repulsion may be resolved by ndings involving the Netrin4 family member. Netrin4 was upregulated in angiogenic endothelium and bound Unc5 receptors, possibly through direct interactions with the coreceptor Neogenin (Lejmi et al. 2008, Qin et al. 2007). Although in vivo mammalian loss-of-function data are lacking, recent in vitro endothelial migration results suggest that both Netrin1

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

and Netrin4 can inhibit VEGF-induced angiogenesis. Conversely, Netrin1- and Netrin4-mediated blood vessel chemoattraction is suggested on the basis of both in vitro vessel proliferation and migration studies and in vivo viral delivery studies. Nevertheless, the cognate in vivo receptors for Netrin1 and Netrin4 remain to be conclusively identied. In addition, these data remain controversial because knockout phenotypes for Unc5b reported by various other groups do not mimic those seen in these studies (Nguyen & Cai 2006, Park et al. 2004, Wilson et al. 2006). Moreover, the functional duality shared by many axon and vessel guidance cues suggests that Netrin/Unc5/DCC may mediate both repulsion and attraction in the vascular system (Yang et al. 2007). Thus, discovering the vascular in vivo receptor(s) for Netrin1 and Netrin4 and the ligand for Unc5b may help to clarify the existing data.

The Ephrin/Eph Family in Vascular Guidance


The Ephrin/Eph receptor tyrosine kinase family of short-range axon guidance molecules was rst identied as chemorepellents (Cheng et al. 1995, Drescher et al. 1995). Their dual functional nature was subsequently elaborated, whereby Ephrin/Eph interactions serve as both attractive and repulsive cues in both the nervous and the vascular systems (Palmer & Klein 2003). Ephrin ligands contain one cupredoxin homolog domain (Figure 3), whereas EphrinA1-5 are anchored to plasma membranes via a glycosyl-phosphatidyl-inositol moiety and EphrinB1-3 have a transmembrane domain followed by a cytoplasmic tail containing a PDZ binding domain that can mediate reverse signaling (Holland et al. 1996). The Eph receptors include 13 family members: EphA1-8, EphB1-4, and EphB6. Eph receptor tyrosine kinases contain one laminin domain, two tumor necrosis factor receptor (TNFR) CRD domains, two Fn3 domains, a transmembrane domain followed by a kinase domain, and a sterile alpha motif (SAM)

domain. In the nervous system, these molecules drive many wiring processes, including midline pathnding, dendritic spine formation, and synaptic plasticity (Palmer & Klein 2003). Ephrin/Eph signaling molecules also control blood vessel development. Ephrin/Eph participate in another key aspect of vascular development: the maintenance of vascular patterning through segregated expression in venous and arteriole vessels. EphB4 is expressed in developing veins, whereas EphrinB2 is found in arteries, providing a key mechanism for repulsive tissue boundary preservation within vascular networks (Adams et al. 1999, Gerety et al. 1999, Wang et al. 1998). These observations are best depicted in ephrinB2 knockout mice, in which angiogenesis defects in both embryonic arteries and veins suggest that both forward and reverse Ephrin/Eph signaling takes place in developing vasculature. Conversely, Ephrin/Eph guidance factors function as positive cues in the context of tumor angiogenesis. For example, EphrinA1 stimulates angiogenesis when expressed on tumor cells, and treatment with the EphA2 receptor ectodomain inhibits neovascularization in these tumor models (Ogawa et al. 2000, Pandey et al. 1995). Therefore, like the other axon guidance family members described, the Ephrin/Eph family of vascular guidance cues also exhibits exquisite dynamic versatility, mediating complex cellular navigational programs within axons and blood vessels as both chemoattractants and repellents. The past decade of discoveries has elucidated key roles for axon guidance molecules in vascular development and has shown that they utilize biological principles similar to those rst observed in the nervous system. These pathways are now a focus of research in the disease setting, for modulating both blood vessel and axonal growth (see sidebar, Axon Guidance Molecules as Targets for Cancer). Having focused on how the vascular and nervous systems share common cues for development, we now turn our attention to the interaction between the vascular and the nervous systems, again with an emphasis on development.
391

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

AXON GUIDANCE MOLECULES AS TARGETS FOR CANCER


As solid tumors begin to grow and become hypoxic, tumor cells recruit blood vessels by secreting VEGF and other angiogenic factors. Blocking angiogenesis is now well established as a mechanism to treat cancer; this process slows tumor growth by reducing nutrient and oxygen transfer to tumor cells (Ferrara & Kerbel 2005). With the discovery of numerous axon guidance molecules playing an essential role in vessel growth during development, investigators have proposed that these mediators of angiogenesis represent a new set of potential cancer targets. Furthermore, several axon guidance molecules have also been studied in the context of tumor cell biology, for example in mediating tumor cell migration. Future studies in both animal models and cancer patients will determine the value of targeting axon guidance molecules as a means to slow tumor growth and disease progression.

vascular network within an even more intricate neural network. Before describing what is known about vascular development in the CNS we rst illustrate in detail the properties of the interface between the vascular and nervous systems, with a focus on the BBB, an essential component of the multicellular NVU.

Molecular Properties of the Blood-Brain Barrier


The now classic experiments demonstrating that a polar dye, trypan blue, injected into blood is specically excluded from the CNS serve as the rst known experimental paradigm in which a barrier between the CNS and peripheral tissue was described (Goldmann 1909, 1913). In the rst set of experiments, investigators prematurely concluded that brain tissue had minimal dye-binding afnity compared with other non-CNS organs. The presence of a BBB was subsequently established with the converse experiment of dye injection into the cerebrospinal uid, the result being staining of brain. Taken together, investigators concluded that various molecules, in this case typan blue (872.88 Da), are uniquely excluded from the CNS and thus that a biological barrier must be in place. The BBB is an essential regulator of bloodbrain exchange of nutrients and waste that operates on several distinct levels (Figure 4). First, endothelial intercellular assemblies such as tight and adherens junctions function as a physical barrier that restricts the paracellular movement of molecules across the BBB.

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

THE INTERFACE BETWEEN THE VASCULAR AND NERVOUS SYSTEMS


The human brain features a large vascular network of 400 miles of blood vessels that receives more than 20% of the bodys cardiac output and utilizes most of its blood glucose (Begley & Brightman 2003). These outstanding characteristics underscore the evolved importance of efcient and thorough exchange of both nutrients and waste between blood and the CNS. This architecture also represents a daunting task for development of a complex


Figure 4 The cellular and molecular properties of the blood-brain barrier (BBB). Many proteins function at the blood-brain barrier (BBB) to regulate molecular exchange. (a) These components include small molecule transporters (P-gp/MRPs, amino-acid, and glucose transporters), large molecule transporters (insulin, LDL, transferrin, and leptin receptors), junctional assemblies (tight and adherens junctions), and metabolic enzymes (P450-related, MAO, and GGT). Tight junctions at the BBB limit most paracellular movement and are composed of several protein families, including occludin, claudin, e-cadherin, ZO, JAM, catenins, cingulin, and actin. (b) Small and/or lipophilic molecules can bypass the tight junctions but are rapidly degraded by intracellular enzymes and actively pumped back into the bloodstream by P-gp and other multidrug-resistant proteins (MRPs). Large molecule, amino-acid, and glucose transporters allow regulated passage of essential nutrients from blood to brain. (c) Electron micrograph of endothelial contacts within the blood-brain barrier (image courtesy of Reese & Karnovsky 1967). Arrows mark intercellular kissing-points indicative of tight junctions.
392 Tam

Watts

BBB: tight junctions, transporters, and transcytosis

a
Capillary lumen (blood)
Endothelial cell P-gp MRPs Amino-acid transporters Glucose transporters Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only. Adherens junctions Tight junctions Large molecule (protein) transport

-GT P450related MAO

Brain

Basement membrane

Barrier regulation: molecular exchange between blood and CNS

Lumenal (blood)
Occludin Claudin E-cadherin ZO JAM Catenins Cingulin Actin

Insulin, LDL, ApoE, transferrin (Tf), leptin

P-gp MRPs Tight junctions

Adherens junctions

Endothelial cell Abluminal (brain)

Small molecules

Ultrastructural image of endothelial tight junctions at the BBB

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

393

Second, several transport mechanisms resident to the BBB jettison unwanted molecules out of the CNS while importing molecules essential for proper brain function. Third, endothelial cells of the BBB express a variety of enzymes that can metabolize unwanted molecules, including toxins (de Boer et al. 2003, Iadecola 2004, Reese & Karnovsky 1967, Rubin & Staddon 1999, Zlokovic 2008). The molecular exchange across the endothelium of the BBB is limited and tightly regulated. Most large, polar metabolites are excluded from blood-brain movement through a complex network of tight junctions and adherens junctions (Figure 4). Integral membrane proteins, such as occludin and claudins, physically associate with the actin cytoskeletal network through zonula occluden adaptor proteins. Claudin-5 is the most well characterized TJ protein within the BBB. It is ubiquitously expressed in all tissue vasculature, and deletion results in leakage of small molecules across the BBB and early postnatal lethality (Nitta et al. 2003). Although cell culture experiments demonstrate that truncated occludin expression functionally compromises the physical barrier because of low transcellular electrical resistance and increased leakage of small molecules across the BBB (Bamforth et al. 1999), occludin is not required for TJ structural formation in vivo (Saitou et al. 2000). Although the BBB is a highly effective gatekeeper, substances that are either lipophilic or smaller than 400 Da may effectively bypass the physical barrier. The metabolic barrier eliminates such chemicals that would otherwise move from the blood into the CNS. As the primary route of drug metabolism, p450-related cytochrome enzyme oxidizes unwanted substances within the cytoplasm of endothelial cells (Alavijeh et al. 2005). Likewise, monoamine oxidase (MAO) also contributes to the metabolic barrier to protect the brain from circulating neurotoxins and biogenic amines. For instance, MAO metabolizes the neurotoxin MPTP and attenuates the parkinsonism associated with this drug (Kalaria & Harik 1987). In some cases, enzymatic activity within the BBB does not
394 Tam

remove unwanted molecules but instead facilitates the transport of essential substances from blood to brain. Through its transpeptidase activity, gamma-glutamyl transpeptidase (GGT) assists in the transfer of amino acids across the BBB (Hawkins et al. 2006). By modulating pyroglutamate levels, GGT indirectly stimulates a wide array of sodium-dependent amino acid transporters at the plasma membrane (Figure 4). Amino acids (AA) represent one of many classes of nutrients essential for proper CNS function and cannot passively diffuse across the BBB because they are neither lipophilic nor smaller than 400 Da. Transport mechanisms tightly facilitate the regulated exchange of these substances across the BBB by utilizing a diverse array of membrane protein carriers and transcytotic receptors. Small metabolites including amino acids, glucose, nucleosides, and vitamins typically move down concentration gradients in the direction from blood to brain through carrier-mediated transporters. GLUT1, a member of the sodium-independent class of glucose carriers, shuttles glucose into the brain and is highly expressed in the BBB. The neurodevelopmental disorder known as GLUT1 deciency syndrome is caused by an autosomal dominant mutation of the glucose transporter and is manifest by seizures and reduced head growth after birth (Wang et al. 2005). These genetic ndings likely underscore the critical importance of proper exchange of nutrients, particularly glucose, across the BBB. The decits observed in humans with this disorder can be modeled in mice that have lost one copy of Glut1 (Wang et al. 2006). Conversely, a subclass of transporters called sodium-dependent AA transporters helps prevent the accumulation of potentially neurotoxic excitatory AAs such as glutamate and aspartate (Zlotnik et al. 2007). Even trace amounts of metabolites that can passively diffuse across the plasma membrane can be highly neuroactive. In these cases, active efux transport mechanisms are necessary to drive molecular exchange against concentration gradients from brain to blood (Figure 4b). The

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

most well-characterized efux conduit is the multidrug resistance protein P-glycoprotein, one of several multidrug resistance-associated proteins (MRPs). This membrane protein efciently removes a diverse structural array of lipophilic molecules through an internal pore (Aller et al. 2009). Researchers propose that through the combined action of several MRPs, both potentially benecial therapeutics and neurotoxic chemicals are rapidly cleared from the brain (Hermann & Bassetti 2007, Loscher & Potschka 2005). Receptor-mediated transcytosis is responsible for the movement of large proteins across the BBB (Pardridge 1988). Leptin, insulin, transferrin, and low-density lipoproteins (LDL) are a few examples of proteins that gain entry into the CNS through binding to their cognate receptors within BBB endothelium and are internalized via endocytic vesicles (Figure 4b). This biological transport is termed receptor-mediated transcytosis. How directionality is achieved when transporting these large proteins across the BBB is not well understood. We presume that the polarity of BBB endothelium and differences between CNS and blood environments drive the transport of these molecules in a direction that benets the organism.

The Neurovascular Unit


For the past two decades, studies have thoroughly established that BBB development, regulation, and maintenance are controlled by various cells in the brain, including pericytes, astrocytes, and neurons (Ek et al. 2006, Janzer & Raff 1987, Stewart & Wiley 1981). The close juxtaposition and physical contact between these different cell types and endothelial cells form the neurovascular unit (NVU) (Figure 5). This functional unit enables efcient oxygen and nutrient ow between various cell types in the CNS, which promotes proper brain function (Banerjee & Bhat 2007, Iadecola 2004). Intercellular communication within the NVU allows the CNS to achieve tight temporal control of cerebral blood ow to

match metabolic needs of the surrounding neural tissue. During development, the neuroepithelium and pericytes actively regulate angiogenesis and vessel guidance. For example, neurons secrete proangiogenic factors such as VEGF (Haigh et al. 2003, Raab et al. 2004) and Wnt ligands (see below) to provide growth and differentiation signals to extending blood vessels. In the mature NVU, neurons further regulate vascular function via signaling through astrocytes, whereas smooth-muscle cells and pericytes control blood ow in response to this neuronal activity. We recognize that cerebral blood ow is regulated by neuronal activity, but the exact mechanisms of NVU function are still under investigation. The functioning of the NVU has been addressed in detail elsewhere (Banerjee & Bhat 2007, Iadecola 2004). Here we address the development of the NVU and its major component, the BBB. In an elegant study utilizing the morphological differences between chick and quail endothelial cells, the unique features of BBB vasculature were shown to be extrinsically induced by the CNS microenvironment (Stewart & Wiley 1981). These studies demonstrated that avascular embryonic quail brain transplanted into chick gut is vascularized by chick peripheral vessels. These vessels take on features of the BBB including tight junctions and the ability to restrict molecular diffusion into the CNS tissue, thus strongly indicating that instructive cues are produced by neuroepithelium to drive barrier formation. It has also become increasingly clear that the barrier is established during the earliest stages of angiogenic development, while astrocytes further drive BBB maturation (Ek et al. 2006, Janzer & Raff 1987). Pericytes and neurons of the NVU are present at the onset of CNS angiogenesis and BBB formation, consistent with the idea that many features of the BBB are initiated during vascular sprouting into the CNS (Virgintino et al. 2007). Furthermore, hallmark BBB components such as Claudin-5 and GLUT1 are present at the earliest stages of CNS angiogenesis; newly formed vessels are established with a physical barrier that can exclude small molecules from entering
395

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

the embryonic CNS (Daneman et al. 2009, Ek et al. 2006). Whereas CNS angiogenesis and BBB formation occur during embryogenesis, formation

of astrocytes and their subsequent ensheathment of vessels start postnatally in rodents, thus the NVU does not fully form until after birth. Detailed rat studies have observed complete

The neurovascular unit (NVU)

a
Astrocyte Synapse

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Pericyte

Astrocyte end-foot Interneuron

Capillary lumen (blood)

Basal lamina

Astrocyte end-foot Astrocyte end-foot Endothelial cell

b
GLUT1 = endothelial cell P-gp = endothelial cell NG2 = pericyte

Collagen IV = basal lamina

GFAP = astrocyte

Collagen IV = basal lamina GFAP = astrocyte

396

Tam

Watts

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

encirclement of vessels by astrocytes three weeks after birth (Caley & Maxwell 1970). Astrocytic regulation of BBB function has been characterized through in vitro coculture experiments and CNS disease studies (Argaw et al. 2009, Hurwitz et al. 1993, Janzer & Raff 1987). However, how astrocytic endfeet affect specic BBB properties during maturation remains to be determined. For example, astrocyte removal had no effect on the physical barrier of the BBB when using the tracer horseradish peroxidase (HRP) to investigate integrity (Krum & Rosenstein 1993). Substantial progress has been made on understanding how the NVU functions in the adult animal (Banerjee & Bhat 2007, Iadecola 2004). Growing evidence also indicates that the dysfunction of the NVU may be a major contributor to neurological disease (Iadecola 2004, Neuwelt et al. 2008, Zlokovic 2008). In particular, altered cerebral blood ow and a compromised BBB have been described in numerous neurodegenerative conditions (see sidebar, The BBB and Neurodegenerative Disease). Nevertheless, to gain further insight into the function of the NVU and associated BBB, a better understanding of the development of these systems must be established.

THE BBB AND NEURODEGENERATIVE DISEASE


In addition to the neuronal loss and inammatory processes that are common hallmarks of neurodegenerative disease, accumulating evidence indicates that the BBB is disrupted in many neurodegenerative diseases, including Alzheimers, Parkinsons, and ALS (Zlokovic 2008). It remains to be seen if the disruption of the BBB is a cause or consequence of neurodegenerative processes. Nevertheless, the fact that the BBB is disrupted is pushing researchers to understand BBB biology in the context of neurodegenerative disease, addressing the following questions, among others: Which molecular mechanisms drive barrier disruption? How does disruption of neurovascular coupling alter CNS function? Is drug delivery altered in neurodegenerative disease? And ultimately, will repairing the BBB slow neurodegenerative disease? This area of research is ripe for discovery and may lead to the development of therapeutics with a primary mechanism of barrier modulation to treat these diseases.

CNS Angiogenesis and BBB Formation: A Role for Wnt Signaling


The canonical Wnt signaling pathway was recently identied as an essential regulator of CNS angiogenesis and BBB development (Daneman et al. 2009, Liebner et al. 2008, Stenman et al. 2008). This developmental

pathway involves Wnt ligand-driven activation of Frizzled and LRP cell surface coreceptors, which then stabilize cellular betacatenin through activated Disheveled protein (Figure 6). The increase in beta-catenin levels then induces transcription of Wnt-responsive genes through Lef-1/TCF DNA binding proteins (van Amerongen & Nusse 2009). In situ hybridization experiments found that the Wnt ligands, wnt7a and wnt7b, are expressed in neural progenitor cells during CNS angiogenesis (Figure 6a). Functionally, Wnt7a/7b were required for developmental angiogenesis induced from the vascular plexus surrounding the spinal cord. Invading vessels utilize


Figure 5 The neurovascular unit (NVU). (a) The mature blood-brain barrier consists of close association and physical contact between endothelial cells, neurons, astrocytic endfeet, basal lamina, and pericytes. The NVU enables efcient cellular communication for proper brain function, such as regulating cerebral blood ow in response to neural activity. (b) Immunohistochemical staining of the cellular components within the mouse NVU. Antibody staining against BBB transporters P-gp and GLUT1 label endothelial cells, whereas NG2 detects the pericytes found along these endothelial cells, which tightly regulate blood ow. Note the tight ensheathment of endothelial cells by the basal lamina (Collagen-IV) and astrocytes (GFAP) visualized by costaining. Astrocyte endfeet (GFAP), in concert with connections from interneurons, regulate neurovascular calcium signaling, blood ow, ionic transport, and water transport.
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 397

a
Perineural vascular endothelium

Embryo neural tube cross-section

Angiogenesis and BBB formation


Endothelial stalk cells BBB endothelial cell differentiation?

Capillary lumen (blood)

Wnts gradient Endothelial tip cell attraction?

Tight junction formation

Neural tube cavity

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Neuroepithelium
Wnts

Nonbrain endothelial cells


Fzd

Brain endothelial cells


Wnt Fzd

LRP

LRP

Inactive dishevelled Active GSK 3b


P

Activated, dishevelled Inactive GSK 3b Axin

Axin Unstable -catenin

APC

APC

Stable -catenin

LEF-1/ TCF

Groucho

Groucho

Wnt-responsive genes off

Transcription of Wnt-responsive genes

Figure 6 Wnts regulate angiogenesis and BBB formation in the nervous system. The canonical Wnt signaling pathway is essential for proper BBB development. (a) Wnt ligands 7a and 7b are specically expressed in neuroepithelium to induce developmental angiogenesis from the perineural vascular plexus into the developing neural tube. These secreted proteins may form attractive gradients that directly shape endothelial tip cell morphology. Furthermore, Wnt signaling in the CNS drives BBB endothelial cell differentiation, thereby inducing glucose transporter-1 (GLUT1) and tight junction protein Claudin-3 expression. (b) The specicity of canonical Wnt signaling on CNS vasculature is highlighted by the observation that Wnt-responsive genes are enriched in BBB endothelium when compared with nonbrain endothelium. This developmental pathway involves Wnt liganddriven activation of Frizzled and LRP cell surface coreceptors, which then activate Dishevelled. This protein inactivates GSK-3 activity in complex with APC and Axin, thereby stabilizing beta-catenin to drive Wnt target genes through Lef-1/TCF DNA binding proteins.
398 Tam

Watts

paracrine signaling derived from the neuroepithelium to coordinate proper BBB development because loss-of-function of either wnt7a/7b in neuroepithelium or of beta-catenin in endothelial cells leads to angiogenic defects and subsequent vascular hemorrhage. These mice exhibit angiogenic defects only in the CNS and not in the periphery, consistent with the observation that Wnt signaling is not activated in non-CNS endothelium (Figure 6b). Although BBB formation parallels angiogenesis in the CNS, proper CNS vascular sprouting alone is not sufcient for complete barrier development. BBB formation requires the expression of a diverse array of gene products to establish barrier function. As previously discussed, GLUT1 is essential for proper brain development because autosomal dominant mutations lead to several neurological maladies and developmental delays (Wang et al. 2005). The tight junction protein Claudin-3, also found predominantly in the BBB vasculature, has been linked to BBB decit under pathological conditions as well (Wolburg et al. 2003). Wnt signaling appears to induce not only CNS angiogenesis, but also expression of BBB components GLUT1 and Claudin-3 (Daneman et al. 2009, Liebner et al. 2008, Stenman et al. 2008). Either Wnt7a/7b or beta-catenin depletion results in a lack of GLUT1 and Claudin-3 expression at the BBB. Conversely, ectopic Wnt7a/7b expression induced GLUT1 expression outside the CNS, whereas addition of Wnt ligands to cultured brain endothelial cells induced both GLUT1 and Claudin-3 protein expression. These ndings emphasize the dual functional nature of Wnt-mediated vascular development in the CNS, whereby both angiogenesis and BBB formation are tightly coupled. We do not yet know if Wnt also plays a role in the nal maturation of the NVU, including driving astrocytic associations with CNS blood vessels. Although the ligands required for Wntdriven BBB development have been at least partially identied as Wnt7a/7b, the cognate receptor(s) have not. Most members of the Frizzled family (Fzd3, 4, 6, 8, 10) and both Lrp5 and Lrp6 are expressed in the CNS.

Daneman and colleagues (2009) speculate that Fzd8 may be important in CNS angiogenesis and BBB development on the basis of its specic enrichment in CNS vasculature. The molecular mechanisms of Wnt-driven BBB development in the CNS could be further elucidated by examining whether endothelial-specic deletion of particular Fzd or LRP isoforms can mimic defects observed in Wnt7a/7b knockout embryos. The specicity of Wnt signaling on CNS vasculature is also highlighted by the observation that canonical Wnt signaling components, such as Lef-1, Tcf7, Axin2, Apcdd1, Ppard, Tbx3, Foxf2, and Stra6, are enriched in BBB endothelium when compared with peripheral tissue endothelium (Daneman et al. 2009). Because this Wnt expression prole was observed in adult mice, the tissue specicity of canonical Wnt signaling appears to be maintained in mature BBB. Could this result indicate that chronic Wnt/beta-catenin signaling is required to maintain proper BBB function within the NVU in the mature animal? The fact that neurons remain a consistent source of Wnt ligand from embryo to adulthood supports such a possibility. For instance, Wnt7a/7b are actively secreted during synaptic activity, which then mediates experience-related regulation of synapse abundance and network complexity (Gogolla et al. 2009). A role for Wnt signaling in BBB maintenance is particularly attractive because it provides a new drug target for both selective opening of the barrier for entry of CNS therapeutics and selective tightening of the BBB in CNS diseases where barrier function may be compromised (Zlokovic 2008). Liebner and colleagues (2008) suggest that Wnt signaling activity is low by the time the BBB is fully mature on the basis of the observation that Wnt signaling activity per unit vessel length shows a precipitous drop after embryonic day 15.5. However, this result could be reective of endothelial cells lengthening dramatically during development, such that the number of nuclei per unit length drops (Liebner et al. 2008). Therefore, BBB function will have to be examined upon suppression of
399

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

Wnt/beta-catenin signaling in the adult animal to determine the validity of this model. In hindsight, the Wnt signaling pathways involvement in BBB development within the CNS should not have come as a surprise. In the developing mouse, Wnt gradients direct axonal pathnding, regulate synaptogenesis, and coordinate axon and dendrite development (Salinas & Zou 2008). Thus the theme of CNS guidance cues regulating vascular development is once again apparent. Several other morphogenic processes including the BMP and hedgehog signaling pathways act in concert with the Wnt pathway in the CNS (Charron & Tessier-Lavigne 2005). Specically, overlapping BMP and Shh ligand gradients cooperate with Netrin1 to drive commissural axons from the roofplate down to the oorplate of the developing spinal cord (Augsburger et al. 1999, Charron et al. 2003). Overlapping Shh and Wnt4 gradients subsequently direct these axonal projections along the anterior/posterior axis to the nal target tissue. Could these signaling pathways be required for proper CNS angiogenesis and BBB development as well? There is precedence for not only shared ligandreceptor paracrine signaling driving both neuronal and vascular patterning, but also Wnt morphogen involvement in CNS angiogenesis and BBB formation, as just described. Indeed, Araya et al. (2008) demonstrated that brain endothelial cells secrete BMP ligands, resulting in astrocyte-derived VEGF production and subsequent BBB breakdown. In addition to the morphogenic signaling pathways, a wide array of axon guidance molecules are also potential candidates for CNS-specic angiogenic and BBB formation cues. Notably, Nrp1 is required for angiogenic sprouting in the CNS (Gerhardt et al. 2004). We need to identify which, if any, of the many axon guidance cues that have been implicated in vessel guidance, branching, development, or maintenance also contribute to CNS angiogenesis and BBB development. The rhodopsin-family G proteincoupled receptor (GPCR), Moody, was recently shown to be required for formation and maintenance
400 Tam

of the Drosophila hemolymph-brain barrier, the functional analog of the mammalian BBB (Bainton et al. 2005). Does GPCR signaling control BBB permeability in mammalian systems as well? Although many GPCRs are expressed in the mouse brain, there are no obvious mammalian orthologs to Moody that can be identied using sequence homology. Nevertheless, it is interesting to note that the adhesion GPCR, gpr125, is upregulated following brain injury and is specically expressed in lung and cortical tissue (Pickering et al. 2008). In addition, the vasodilator bradykinin acutely modulates BBB permeability through its cognate GPCRs, bdkrb1 and b2 (Bartus et al. 1996, Su et al. 2009). We are only just beginning to understand the signaling pathways that coordinate the formation of a fully functional BBB. Furthermore, specic pathways may drive BBB development within different regions of the CNS. Consistent with this possibility, some evidence indicates that Wnt/beta-catenin signaling is not necessary for pan-CNS angiogenesis and BBB formation. For instance, Wnt7a/7b ligands are not expressed in dorsal neural tubes, and as a consequence, Wnt7a/7b knockout mice can still form proper blood vessels in this region (Stenman et al. 2008). Similarly, beta-catenin knockouts exhibit normal BBB formation in the hindbrain (Daneman et al. 2009), and Fzd4 knockout mice show barrier decits in the cerebellum, but not the cortex, suggesting that multiple Fzd receptors may coordinate BBB development in a region-specic fashion (Ye et al. 2009). Therefore, unique positions within the CNS seem to require activation of other signaling pathways. Unbiased approaches toward uncovering novel molecular mechanisms of BBB development and maintenance are needed at this juncture. Signaling pathways coregulated with Wnt activation in CNS endothelium may suggest participation in BBB formation. Downstream of Wnt signaling, many of the BBB-related genes directly or indirectly regulated by Wnt/beta-catenin have yet to be identied. Investigation of BBB endothelial cell expression

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Watts

proles along the developmental timeline may paint a more comprehensive picture of BBB development, maturation, and maintenance.

CONCLUSIONS AND FUTURE DIRECTIONS


Mirroring the sequential origin of the nervous and vascular systems, researchers have applied principles of nervous system development to address vascular biology questions. This approach stemmed from the original observation that the nervous and vascular systems share anatomical and cellular similarities. Moreover, both systems must be present throughout an organism to deliver and receive vital information and nutrients. It is apparent now that similar molecular mechanisms drive both vascular and neuronal network formation. These discoveries have been key to accelerating our understanding of how blood vessels develop. Also, the vascular system can directly impact nervous system development and vice versa; thus caution should be taken when characterizing phenotypes after disrupting signaling pathways in either system. We have described examples of specic axon guidance molecules regulating vascular biology. Although not comprehensive, these examples illustrate the inuence that discoveries in the eld of axon guidance have had on understanding vascular development. However, much remains unknown. For instance, the role that VEGF plays in neural development and function is just beginning to be unraveled (Ruiz de Almodovar et al. 2009). In addition, we have not addressed in this review the close relationship between blood vessels and neural progenitor cells at the vascular niche (Barami 2008). Many important questions remain that would further our understanding of the relationship between vascular and nervous system development. Are there other guidance cues that mediate both vascular and neural development? How are developmentally sequential events, such as initial peripheral blood vessel innervation followed by neuronal process innervation, regulated? Are molecular mechanisms of vascular and neuronal remodeling also shared

or codependent? How did the vascular system co-opt nervous system cues? What role do these common cues play in pathological angiogenesis or neurological disease? After more than a decade of applying insight from neuronal development paradigms to the vascular system, it is clear that this path of investigation is yielding key ndings. Much less is understood about the development, maturation, and maintenance of the blood-brain barrier (BBB). Understanding how the BBB develops and functions is of enormous importance. This need is fueled by the fact that the barrier hinders therapeutic treatment of neurological diseases and because it may also be at the center of neurodegenerative disease biology. Subsequent to the original discovery that the CNS microenvironment directs blood vessel differentiation to form the BBB (Stewart & Wiley 1981), the recent identication of the Wnt/beta-catenin signaling pathway regulating CNS angiogenesis and BBB formation is likely just the rst of many examples of regulatory cues derived from the CNS that drive brain endothelial cell development and differentiation. A unique Frizzled receptor ligand, Norrin, has recently been described to act via Fzd4, LRP5, and a tetraspanin molecule, TSPAN12, to regulate retinal vascular development specically (Junge et al. 2009, Ye et al. 2009). In addition, Norrin, Fzd4, Lrp5, and Tspan12 knockout mice show a vascular hemorrhage phenotype in the retina, consistent with the disruption of the retinal-blood barrier. These data suggest that unique Wnt signaling pathways have evolved in a regional-specic fashion to coordinate endothelial cell differentiation and barrier formation. Beyond Wnt signaling in CNS angiogenesis and BBB formation, many questions remain about how the BBB forms, matures, and is maintained. The barrier also consists of other cell types, including pericytes, astrocytes, and neuronal components. How is this complex NVU formed? How is the barrier actively maintained in the adult animal? What are the roles of the various cellular components in the development, maturation, and maintenance
401

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

www.annualreviews.org Angiogenesis and the Blood-Brain Barrier

of the BBB? Discovering the molecular components that address these broader questions will deepen our understanding of the BBB, thus enabling researchers to address unresolved issues of drug penetration into the CNS

and barrier disruption in neurological disease paradigms. Using recent history as an example, clues to crack the mystery of BBB formation may be derived from applying principles of neuronal development to the vascular system.

DISCLOSURE STATEMENT
The authors are employees of Genentech, Inc.

ACKNOWLEDGMENTS
Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

We apologize to our colleagues whose research we could not cite owing to space limitations. We thank Allison Bruce for the generation of illustrations and thank Kimberly Scearce-Levie, Weilan Ye, Joy Yu, Morgan Sheng, and Marc Tessier-Lavigne for their helpful comments on the manuscript. LITERATURE CITED
Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, et al. 1999. Roles of ephrinB ligands and EphB receptors in cardiovascular development: demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes Dev. 13:295306 Alavijeh MS, Chishty M, Qaiser MZ, Palmer AM. 2005. Drug metabolism and pharmacokinetics, the bloodbrain barrier, and central nervous system drug discovery. NeuroRx 2:55471 Aller SG, Yu J, Ward A, Weng Y, Chittaboina S, et al. 2009. Structure of P-glycoprotein reveals a molecular basis for poly-specic drug binding. Science 323:171822 Appleton BA, Wu P, Maloney J, Yin J, Liang WC, et al. 2007. Structural studies of neuropilin/antibody complexes provide insights into semaphorin and VEGF binding. EMBO J. 26:490212 Araya R, Kudo M, Kawano M, Ishii K, Hashikawa T, et al. 2008. BMP signaling through BMPRIA in astrocytes is essential for proper cerebral angiogenesis and formation of the blood-brain-barrier. Mol. Cell Neurosci. 38:41730 Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. 2009. VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proc. Natl. Acad. Sci. USA 106:197782 Augsburger A, Schuchardt A, Hoskins S, Dodd J, Butler S. 1999. BMPs as mediators of roof plate repulsion of commissural neurons. Neuron 24:12741 Bainton RJ, Tsai LT, Schwabe T, DeSalvo M, Gaul U, Heberlein U. 2005. moody encodes two GPCRs that regulate cocaine behaviors and blood-brain barrier permeability in Drosophila. Cell 123:14556 Bamforth SD, Kniesel U, Wolburg H, Engelhardt B, Risau W. 1999. A dominant mutant of occludin disrupts tight junction structure and function. J. Cell Sci. 112(Pt. 12):187988 Banerjee S, Bhat MA. 2007. Neuron-glial interactions in blood-brain barrier formation. Annu. Rev. Neurosci. 30:23558 Barami K. 2008. Relationship of neural stem cells with their vascular niche: implications in the malignant progression of gliomas. J. Clin. Neurosci. 15:119397 Barres BA. 2008. The mystery and magic of glia: a perspective on their roles in health and disease. Neuron 60:43040 Bartus RT, Elliott P, Hayward N, Dean R, McEwen EL, Fisher SK. 1996. Permeability of the blood brain barrier by the bradykinin agonist, RMP-7: evidence for a sensitive, auto-regulated, receptor-mediated system. Immunopharmacology 33:27078 Bedell VM, Yeo SY, Park KW, Chung J, Seth P, et al. 2005. roundabout4 is essential for angiogenesis in vivo. Proc. Natl. Acad. Sci. USA 102:637378 Begley DJ, Brightman MW. 2003. Structural and functional aspects of the blood-brain barrier. Prog. Drug Res. 61:3978
402 Tam

Watts

Bergers G, Song S. 2005. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol. 7:45264 Betsholtz C, Lindblom P, Gerhardt H. 2005. Role of pericytes in vascular morphogenesis. Exs 2005:11525 Bouvr ee K, Larriv ee B, Lv X, Yuan L, DeLafarge B, et al. 2008. Netrin-1 inhibits sprouting angiogenesis in developing avian embryos. Dev. Biol. 318:17283 Butler SJ, Dodd J. 2003. A role for BMP heterodimers in roof plate-mediated repulsion of commissural axons. Neuron 38:389401 Caley DW, Maxwell DS. 1970. Development of the blood vessels and extracellular spaces during postnatal maturation of rat cerebral cortex. J. Comp. Neurol. 138:3147 Carmeliet P, Tessier-Lavigne M. 2005. Common mechanisms of nerve and blood vessel wiring. Nature 436:193200 Charron F, Stein E, Jeong J, McMahon AP, Tessier-Lavigne M. 2003. The morphogen sonic hedgehog is an axonal chemoattractant that collaborates with netrin-1 in midline axon guidance. Cell 113:1123 Charron F, Tessier-Lavigne M. 2005. Novel brain wiring functions for classical morphogens: a role as graded positional cues in axon guidance. Development 132:225162 Chen H, Ch edotal A, He Z, Goodman CS, Tessier-Lavigne M. 1997. Neuropilin-2, a novel member of the neuropilin family, is a high afnity receptor for the semaphorins Sema E and Sema IV but not Sema III. Neuron 19:54759 Cheng HJ, Bagri A, Yaron A, Stein E, Pleasure SJ, Tessier-Lavigne M. 2001. Plexin-A3 mediates semaphorin signaling and regulates the development of hippocampal axonal projections. Neuron 32:24963 Cheng HJ, Nakamoto M, Bergemann AD, Flanagan JG. 1995. Complementary gradients in expression and binding of ELF-1 and Mek4 in development of the topographic retinotectal projection map. Cell 82:371 81 Cho NK, Keyes L, Johnson E, Heller J, Ryner L, et al. 2002. Developmental control of blood cell migration by the Drosophila VEGF pathway. Cell 108:86576 Coultas L, Chawengsaksophak K, Rossant J. 2005. Endothelial cells and VEGF in vascular development. Nature 438:93745 Daneman R, Agalliu D, Zhou L, Kuhnert F, Kuo CJ, Barres BA. 2009. Wnt/beta-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc. Natl. Acad. Sci. USA 106:64146 de Boer AG, van der Sandt IC, Gaillard PJ. 2003. The role of drug transporters at the blood-brain barrier. Annu. Rev. Pharmacol. Toxicol. 43:62956 de Castro F, Lopez-Mascaraque L, De Carlos JA. 2007. Cajal: lessons on brain development. Brain Res. Rev. 55:48189 Dickson BJ. 2002. Molecular mechanisms of axon guidance. Science 298:195964 Dickson BJ, Gilestro GF. 2006. Regulation of commissural axon pathnding by slit and its Robo receptors. Annu. Rev. Cell Dev. Biol. 22:65175 Drescher U, Kremoser C, Handwerker C, Loschinger J, Noda M, Bonhoeffer F. 1995. In vitro guidance of retinal ganglion cell axons by RAGS, a 25 kDa tectal protein related to ligands for Eph receptor tyrosine kinases. Cell 82:35970 Duarte A, Hirashima M, Benedito R, Trindade A, Diniz P, et al. 2004. Dosage-sensitive requirement for mouse Dll4 in artery development. Genes Dev. 18:247478 Ek CJ, Dziegielewska KM, Stolp H, Saunders NR. 2006. Functional effectiveness of the blood-brain barrier to small water-soluble molecules in developing and adult opossum (Monodelphis domestica). J. Comp. Neurol. 496:1326 Fazeli A, Dickinson SL, Hermiston ML, Tighe RV, Steen RG, et al. 1997. Phenotype of mice lacking functional Deleted in colorectal cancer (Dcc) gene. Nature 386:796804 Ferrara N, Kerbel RS. 2005. Angiogenesis as a therapeutic target. Nature 438:96774 Fisher SA, Burggren WW. 2007. Role of hypoxia in the evolution and development of the cardiovascular system. Antioxid. Redox Signal. 9:133952 Fong GH. 2009. Regulation of angiogenesis by oxygen sensing mechanisms. J. Mol. Med. 87:54960 Fujisawa H. 2004. Discovery of semaphorin receptors, neuropilin and plexin, and their functions in neural development. J. Neurobiol. 59:2433
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 403

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Gaiano N, Fishell G. 2002. The role of notch in promoting glial and neural stem cell fates. Annu. Rev. Neurosci. 25:47190 Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, et al. 2004. Haploinsufciency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc. Natl. Acad. Sci. USA 101:1594954 Gerety SS, Wang HU, Chen ZF, Anderson DJ. 1999. Symmetrical mutant phenotypes of the receptor EphB4 and its specic transmembrane ligand ephrin-B2 in cardiovascular development. Mol. Cell 4:40314 Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, et al. 2003. VEGF guides angiogenic sprouting utilizing endothelial tip cell lopodia. J. Cell Biol. 161:116377 Gerhardt H, Ruhrberg C, Abramsson A, Fujisawa H, Shima D, Betsholtz C. 2004. Neuropilin-1 is required for endothelial tip cell guidance in the developing central nervous system. Dev. Dyn. 231:5039 Ghabrial AS, Krasnow MA. 2006. Social interactions among epithelial cells during tracheal branching morphogenesis. Nature 441:74649 Gitler AD, Lu MM, Epstein JA. 2004. PlexinD1 and semaphorin signaling are required in endothelial cells for cardiovascular development. Dev. Cell 7:10716 Gogolla N, Galimberti I, Deguchi Y, Caroni P. 2009. Wnt signaling mediates experience-related regulation of synapse numbers and mossy ber connectivities in the adult hippocampus. Neuron 62:51025 Goldmann EE. 1909. Die a ussere und innere sekretion des gesunden und kranken organismus im lichte der vitalen Farbung. Beitr. Klin. Chir. 64:192265 Goldmann EE. 1913. Vitalfarbung am zentralnervensystem. Abh. Preuss. Akad. Wiss. Phys.-Math 1:160 Golgi C. 1906. The neuron doctrinetheory and facts. http://nobelprize.org/nobel_prizes/medicine/laureates/ 1906/golgi-lecture.pdf Gu C, Limberg BJ, Whitaker GB, Perman B, Leahy DJ, et al. 2002. Characterization of neuropilin-1 structural features that confer binding to semaphorin 3A and vascular endothelial growth factor 165. J. Biol. Chem. 277:1806976 Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, et al. 2003. Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Dev. Cell 5:4557 Gu C, Yoshida Y, Livet J, Reimert DV, Mann F, et al. 2005. Semaphorin 3E and plexin-D1 control vascular pattern independently of neuropilins. Science 307:26568 Haigh JJ, Morelli PI, Gerhardt H, Haigh K, Tsien J, et al. 2003. Cortical and retinal defects caused by dosage-dependent reductions in VEGF-A paracrine signaling. Dev. Biol. 262:22541 Hawkins BT, Davis TP. 2005. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol. Rev. 57:17385 Hawkins RA, OKane RL, Simpson IA, Vina JR. 2006. Structure of the blood-brain barrier and its role in the transport of amino acids. J. Nutr. 136:S21826 He Z, Tessier-Lavigne M. 1997. Neuropilin is a receptor for the axonal chemorepellent Semaphorin III. Cell 90:73951 Hellstrom M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, et al. 2007. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 445:77680 Hermann DM, Bassetti CL. 2007. Implications of ATP-binding cassette transporters for brain pharmacotherapies. Trends Pharmacol. Sci. 28:12834 Holland SJ, Gale NW, Mbamalu G, Yancopoulos GD, Henkemeyer M, Pawson T. 1996. Bidirectional signalling through the EPH-family receptor Nuk and its transmembrane ligands. Nature 383:72225 Hong K, Hinck L, Nishiyama M, Poo MM, Tessier-Lavigne M, Stein E. 1999. A ligand-gated association between cytoplasmic domains of UNC5 and DCC family receptors converts netrin-induced growth cone attraction to repulsion. Cell 97:92741 Honma Y, Araki T, Gianino S, Bruce A, Heuckeroth R, et al. 2002. Artemin is a vascular-derived neurotropic factor for developing sympathetic neurons. Neuron 35:26782 Huminiecki L, Gorn M, Suchting S, Poulsom R, Bicknell R. 2002. Magic roundabout is a new member of the roundabout receptor family that is endothelial specic and expressed at sites of active angiogenesis. Genomics 79:54752 Hurwitz AA, Berman JW, Rashbaum WK, Lyman WD. 1993. Human fetal astrocytes induce the expression of blood-brain barrier specic proteins by autologous endothelial cells. Brain Res. 625:23843
404 Tam

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Watts

Iadecola C. 2004. Neurovascular regulation in the normal brain and in Alzheimers disease. Nat. Rev. Neurosci. 5:34760 Ishii N, Wadsworth WG, Stern BD, Culotti JG, Hedgecock EM. 1992. UNC-6, a laminin-related protein, guides cell and pioneer axon migrations in C. elegans. Neuron 9:87381 Janzer RC, Raff MC. 1987. Astrocytes induce blood-brain barrier properties in endothelial cells. Nature 325:25357 Jones EA, Yuan L, Breant C, Watts RJ, Eichmann A. 2008. Separating genetic and hemodynamic defects in neuropilin 1 knockout embryos. Development 135:247988 Junge HJ, Yang S, Burton JB, Paes K, Shu X, et al. 2009. TSPAN12 regulates retinal vascular development by promoting Norrin- but not Wnt-induced FZD4/beta-catenin signaling. Cell 139:299311 Kalaria RN, Harik SI. 1987. Blood-brain barrier monoamine oxidase: enzyme characterization in cerebral microvessels and other tissues from six mammalian species, including human. J. Neurochem. 49:85664 Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M, et al. 1999. A requirement for neuropilin-1 in embryonic vessel formation. Development 126:4895902 Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, et al. 1989. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science 246:130912 Keino-Masu K, Masu M, Hinck L, Leonardo ED, Chan SS, et al. 1996. Deleted in Colorectal Cancer (DCC) encodes a netrin receptor. Cell 87:17585 Keleman K, Dickson BJ. 2001. Short- and long-range repulsion by the Drosophila Unc5 netrin receptor. Neuron 32:60517 Kennedy TE, Serani T, de la Torre JR, Tessier-Lavigne M. 1994. Netrins are diffusible chemotropic factors for commissural axons in the embryonic spinal cord. Cell 78:42535 Kidd T, Bland KS, Goodman CS. 1999. Slit is the midline repellent for the robo receptor in Drosophila. Cell 96:78594 Kidd T, Brose K, Mitchell KJ, Fetter RD, Tessier-Lavigne M, et al. 1998. Roundabout controls axon crossing of the CNS midline and denes a novel subfamily of evolutionarily conserved guidance receptors. Cell 92:20515 Kolodkin AL. 1996. Growth cones and the cues that repel them. Trends Neurosci. 19:50713 Kolodkin AL, Levengood DV, Rowe EG, Tai YT, Giger RJ, Ginty DD. 1997. Neuropilin is a semaphorin III receptor. Cell 90:75362 Kolodkin AL, Matthes DJ, OConnor TP, Patel NH, Admon A, et al. 1992. Fasciclin IV: sequence, expression, and function during growth cone guidance in the grasshopper embryo. Neuron 9:83145 Krebs LT, Shutter JR, Tanigaki K, Honjo T, Stark KL, Gridley T. 2004. Haploinsufcient lethality and formation of arteriovenous malformations in Notch pathway mutants. Genes Dev. 18:246973 Krum JM, Rosenstein JM. 1993. Effect of astroglial degeneration on the blood-brain barrier to protein in neonatal rats. Brain Res. Dev. Brain Res. 74:4150 Larriv ee B, Freitas C, Suchting S, Brunet I, Eichmann A. 2009. Guidance of vascular development: lessons from the nervous system. Circ. Res. 104:42841 Larriv ee B, Freitas C, Trombe M, Lv X, Delafarge B, et al. 2007. Activation of the UNC5B receptor by Netrin-1 inhibits sprouting angiogenesis. Genes Dev. 21:243347 Lawson ND, Weinstein BM. 2002. In vivo imaging of embryonic vascular development using transgenic zebrash. Dev. Biol. 248:30718 Lejmi E, Leconte L, Pedron-Mazoyer S, Ropert S, Raoul W, et al. 2008. Netrin-4 inhibits angiogenesis via binding to neogenin and recruitment of Unc5B. Proc. Natl. Acad. Sci. USA 105:1249196 Leonardo ED, Hinck L, Masu M, Keino-Masu K, Ackerman SL, Tessier-Lavigne M. 1997. Vertebrate homologues of C. elegans UNC-5 are candidate netrin receptors. Nature 386:83338 Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. 1989. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246:13069 Liebner S, Corada M, Bangsow T, Babbage J, Taddei A, et al. 2008. Wnt/beta-catenin signaling controls development of the blood-brain barrier. J. Cell Biol. 183:40917 Lobov IB, Renard RA, Papadopoulos N, Gale NW, Thurston G, et al. 2007. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc. Natl. Acad. Sci. USA 104:3219 24
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 405

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Loscher W, Potschka H. 2005. Drug resistance in brain diseases and the role of drug efux transporters. Nat. Rev. Neurosci. 6:591602 Lowery LA, Van Vactor D. 2009. The trip of the tip: understanding the growth cone machinery. Nat. Rev. Mol. Cell Biol. 10:33243 Lu X, Le Noble F, Yuan L, Jiang Q, De Lafarge B, et al. 2004. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 432:17986 Lubarsky B, Krasnow MA. 2003. Tube morphogenesis: making and shaping biological tubes. Cell 112:19 28 Lyuksyutova AI, Lu CC, Milanesio N, King LA, Guo N, et al. 2003. Anterior-posterior guidance of commissural axons by Wnt-frizzled signaling. Science 302:198488 Makita T, Sucov HM, Gariepy CE, Yanagisawa M, Ginty DD. 2008. Endothelins are vascular-derived axonal guidance cues for developing sympathetic neurons. Nature 452:75963 Miao HQ, Soker S, Feiner L, Alonso JL, Raper JA, Klagsbrun M. 1999. Neuropilin-1 mediates collapsin1/semaphorin III inhibition of endothelial cell motility: functional competition of collapsin-1 and vascular endothelial growth factor-165. J. Cell Biol. 146:23342 Miller G. 2009. Origins. On the origin of the nervous system. Science 325:2426 Moore SW, Tessier-Lavigne M, Kennedy TE. 2007. Netrins and their receptors. Adv. Exp. Med. Biol. 621:17 31 Mukouyama YS, Gerber HP, Ferrara N, Gu C, Anderson DJ. 2005. Peripheral nerve-derived VEGF promotes arterial differentiation via neuropilin 1-mediated positive feedback. Development 132:94152 Mukouyama YS, Shin D, Britsch S, Taniguchi M, Anderson DJ. 2002. Sensory nerves determine the pattern of arterial differentiation and blood vessel branching in the skin. Cell 109:693705 Neuwelt E, Abbott NJ, Abrey L, Banks WA, Blakley B, et al. 2008. Strategies to advance translational research into brain barriers. Lancet Neurol. 7:8496 Nguyen A, Cai H. 2006. Netrin-1 induces angiogenesis via a DCC-dependent ERK1/2-eNOS feed-forward mechanism. Proc. Natl. Acad. Sci. USA 103:653035 Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, et al. 2003. Size-selective loosening of the blood-brain barrier in claudin-5-decient mice. J. Cell Biol. 161:65360 ODonnell M, Chance RK, Bashaw GJ. 2009. Axon growth and guidance: receptor regulation and signal transduction. Annu. Rev. Neurosci. 32:383412 Ogawa K, Pasqualini R, Lindberg RA, Kain R, Freeman AL, Pasquale EB. 2000. The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19:604352 Palmer A, Klein R. 2003. Multiple roles of ephrins in morphogenesis, neuronal networking, and brain function. Genes Dev. 17:142950 Pan Q, Chanthery Y, Liang WC, Stawicki S, Mak J, et al. 2007. Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell 11:5367 Pandey A, Shao H, Marks RM, Polverini PJ, Dixit VM. 1995. Role of B61, the ligand for the Eck receptor tyrosine kinase, in TNF-alpha-induced angiogenesis. Science 268:56769 Pardridge WM. 1988. Recent advances in blood-brain barrier transport. Annu. Rev. Pharmacol. Toxicol. 28:25 39 Park KW, Crouse D, Lee M, Karnik SK, Sorensen LK, et al. 2004. The axonal attractant Netrin-1 is an angiogenic factor. Proc. Natl. Acad. Sci. USA 101:1621015 Park KW, Morrison CM, Sorensen LK, Jones CA, Rao Y, et al. 2003. Robo4 is a vascular-specic receptor that inhibits endothelial migration. Dev. Biol. 261:25167 Pickering C, Hagglund M, Szmydynger-Chodobska J, Marques F, Palha JA, et al. 2008. The Adhesion GPCR GPR125 is specically expressed in the choroid plexus and is upregulated following brain injury. BMC Neurosci. 9:97 Qin S, Yu L, Gao Y, Zhou R, Zhang C. 2007. Characterization of the receptors for axon guidance factor netrin-4 and identication of the binding domains. Mol. Cell Neurosci. 34:24350 Raab S, Beck H, Gaumann A, Yuce A, Gerber HP, et al. 2004. Impaired brain angiogenesis and neuronal apoptosis induced by conditional homozygous inactivation of vascular endothelial growth factor. Thromb. Haemost. 91:595605
406 Tam

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Watts

Ramon y Cajal S. 1906. The structure and connexions of neurons. http://nobelprize.org/nobel_prizes/ medicine/laureates/1906/cajal-lecture.pdf Ramon y Cajal S. 1917. Recuerdos de mi vida. Madrid: Moya Raper JA. 2000. Semaphorins and their receptors in vertebrates and invertebrates. Curr. Opin. Neurobiol. 10:8894 Reese TS, Karnovsky MJ. 1967. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J. Cell Biol. 34:20717 Rossignol M, Gagnon ML, Klagsbrun M. 2000. Genomic organization of human neuropilin-1 and neuropilin2 genes: identication and distribution of splice variants and soluble isoforms. Genomics 70:21122 Rubin LL, Staddon JM. 1999. The cell biology of the blood-brain barrier. Annu. Rev. Neurosci. 22:1128 Ruhrberg C, Gerhardt H, Golding M, Watson R, Ioannidou S, et al. 2002. Spatially restricted patterning cues provided by heparin-binding VEGF-A control blood vessel branching morphogenesis. Genes Dev. 16:268498 Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P. 2009. Role and therapeutic potential of VEGF in the nervous system. Physiol. Rev. 89:60748 Saitou M, Furuse M, Sasaki H, Schulzke JD, Fromm M, et al. 2000. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol. Biol. Cell 11:413142 Salinas PC, Zou Y. 2008. Wnt signaling in neural circuit assembly. Annu. Rev. Neurosci. 31:33958 Salminen M, Meyer BI, Bober E, Gruss P. 2000. Netrin 1 is required for semicircular canal formation in the mouse inner ear. Development 127:1322 Seeger M, Tear G, Ferres-Marco D, Goodman CS. 1993. Mutations affecting growth cone guidance in Drosophila: genes necessary for guidance toward or away from the midline. Neuron 10:40926 Serani T, Colamarino SA, Leonardo ED, Wang H, Beddington R, et al. 1996. Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system. Cell 87:100114 Serani T, Kennedy TE, Galko MJ, Mirzayan C, Jessell TM, Tessier-Lavigne M. 1994. The netrins dene a family of axon outgrowth-promoting proteins homologous to C. elegans UNC-6. Cell 78:40924 Serini G, Valdembri D, Zanivan S, Morterra G, Burkhardt C, et al. 2003. Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 424:39197 Sheldon H, Andre M, Legg JA, Heal P, Herbert JM, et al. 2009. Active involvement of Robo1 and Robo4 in lopodia formation and endothelial cell motility mediated via WASP and other actin nucleationpromoting factors. FASEB J. 23:51322 Shoji W, Isogai S, Sato-Maeda M, Obinata M, Kuwada JY. 2003. Semaphorin3a1 regulates angioblast migration and vascular development in zebrash embryos. Development 130:322736 Soker S, Miao HQ, Nomi M, Takashima S, Klagsbrun M. 2002. VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J. Cell Biochem. 85:357 68 Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. 1998. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specic receptor for vascular endothelial growth factor. Cell 92:735 45 Song H, Ming G, He Z, Lehmann M, McKerracher L, et al. 1998. Conversion of neuronal growth cone responses from repulsion to attraction by cyclic nucleotides. Science 281:151518 Sperry RW. 1963. Chemoafnity in the orderly growth of nerve ber patterns and connections. Proc. Natl. Acad. Sci. USA 50:70310 Stein E, Tessier-Lavigne M. 2001. Hierarchical organization of guidance receptors: silencing of netrin attraction by slit through a Robo/DCC receptor complex. Science 291:192838 Stenman JM, Rajagopal J, Carroll TJ, Ishibashi M, McMahon J, McMahon AP. 2008. Canonical Wnt signaling regulates organ-specic assembly and differentiation of CNS vasculature. Science 322:124750 Stewart PA, Wiley MJ. 1981. Developing nervous tissue induces formation of blood-brain barrier characteristics in invading endothelial cells: a study using quailchick transplantation chimeras. Dev. Biol. 84:18392 Su J, Cui M, Tang Y, Zhou H, Liu L, Dong Q. 2009. Blockade of bradykinin B2 receptor more effectively reduces postischemic blood-brain barrier disruption and cytokines release than B1 receptor inhibition. Biochem. Biophys. Res. Commun. 388:20511
www.annualreviews.org Angiogenesis and the Blood-Brain Barrier 407

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Suchting S, Freitas C, le Noble F, Benedito R, Breant C, et al. 2007. The Notch ligand Delta-like 4 negatively regulates endothelial tip cell formation and vessel branching. Proc. Natl. Acad. Sci. USA 104:322530 Suchting S, Heal P, Tahtis K, Stewart LM, Bicknell R. 2005. Soluble Robo4 receptor inhibits in vivo angiogenesis and endothelial cell migration. FASEB J. 19:12123 Takashima S, Kitakaze M, Asakura M, Asanuma H, Sanada S, et al. 2002. Targeting of both mouse neuropilin1 and neuropilin-2 genes severely impairs developmental yolk sac and embryonic angiogenesis. Proc. Natl. Acad. Sci. USA 99:365762 Tamagnone L, Artigiani S, Chen H, He Z, Ming GI, et al. 1999. Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell 99:7180 Tessier-Lavigne M, Goodman CS. 1996. The molecular biology of axon guidance. Science 274:112333 Torres-Vazquez J, Gitler AD, Fraser SD, Berk JD, Van NP, et al. 2004. Semaphorin-plexin signaling guides patterning of the developing vasculature. Dev. Cell 7:11723 Tran TS, Kolodkin AL, Bharadwaj R. 2007. Semaphorin regulation of cellular morphology. Annu. Rev. Cell Dev. Biol. 23:26392 van Amerongen R, Nusse R. 2009. Towards an integrated view of Wnt signaling in development. Development 136:320514 van der Zwaag B, Hellemons AJ, Leenders WP, Burbach JP, Brunner HG, et al. 2002. PLEXIN-D1, a novel plexin family member, is expressed in vascular endothelium and the central nervous system during mouse embryogenesis. Dev. Dyn. 225:33643 Vesalius A. 1543. De Humani Corporis Fabrica. Basel: Oporinus Vieira JM, Schwarz Q, Ruhrberg C. 2007. Selective requirements for NRP1 ligands during neurovascular patterning. Development 134:183343 Virgintino D, Girolamo F, Errede M, Capobianco C, Robertson D, et al. 2007. An intimate interplay between precocious, migrating pericytes and endothelial cells governs human fetal brain angiogenesis. Angiogenesis 10:3545 Wang B, Xiao Y, Ding BB, Zhang N, Yuan X, et al. 2003. Induction of tumor angiogenesis by Slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell 4:1929 Wang D, Pascual JM, Yang H, Engelstad K, Jhung S, et al. 2005. Glut-1 deciency syndrome: clinical, genetic, and therapeutic aspects. Ann. Neurol. 57:11118 Wang D, Pascual JM, Yang H, Engelstad K, Mao X, et al. 2006. A mouse model for Glut-1 haploinsufciency. Hum. Mol. Genet. 15:116979 Wang HU, Chen ZF, Anderson DJ. 1998. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 93:74153 Wang KH, Brose K, Arnott D, Kidd T, Goodman CS, et al. 1999. Biochemical purication of a mammalian slit protein as a positive regulator of sensory axon elongation and branching. Cell 96:77184 Wilson BD, Ii M, Park KW, Suli A, Sorensen LK, et al. 2006. Netrins promote developmental and therapeutic angiogenesis. Science 313:64044 Wolburg H, Wolburg-Buchholz K, Kraus J, Rascher-Eggstein G, Liebner S, et al. 2003. Localization of claudin-3 in tight junctions of the blood-brain barrier is selectively lost during experimental autoimmune encephalomyelitis and human glioblastoma multiforme. Acta Neuropathol. 105:58692 Yang Y, Zou L, Wang Y, Xu KS, Zhang JX, Zhang JH. 2007. Axon guidance cue Netrin-1 has dual function in angiogenesis. Cancer Biol. Ther. 6:74348 Ye X, Wang Y, Cahill H, Yu M, Badea TC, et al. 2009. Norrin, frizzled-4, and Lrp5 signaling in endothelial cells controls a genetic program for retinal vascularization. Cell 139:28598 Yoshikawa S, McKinnon RD, Kokel M, Thomas JB. 2003. Wnt-mediated axon guidance via the Drosophila Derailed receptor. Nature 422:58388 Zallen JA, Yi BA, Bargmann CI. 1998. The conserved immunoglobulin superfamily member SAX-3/Robo directs multiple aspects of axon guidance in C. elegans. Cell 92:21727 Zlokovic BV. 2008. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57:178201 Zlotnik A, Gurevich B, Tkachov S, Maoz I, Shapira Y, Teichberg VI. 2007. Brain neuroprotection by scavenging blood glutamate. Exp. Neurol. 203:21320

Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

408

Tam

Watts

Annual Review of Neuroscience

Contents
Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Volume 33, 2010

Attention, Intention, and Priority in the Parietal Lobe James W. Bisley and Michael E. Goldberg p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1 The Subplate and Early Cortical Circuits Patrick O. Kanold and Heiko J. Luhmann p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 23 Fly Motion Vision Alexander Borst, Juergen Haag, and Dierk F. Reiff p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 49 Molecular Pathways of Frontotemporal Lobar Degeneration Kristel Sleegers, Marc Cruts, and Christine Van Broeckhoven p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 71 Error Correction, Sensory Prediction, and Adaptation in Motor Control Reza Shadmehr, Maurice A. Smith, and John W. Krakauer p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 89 How Does Neuroscience Affect Our Conception of Volition? Adina L. Roskies p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 109 Watching Synaptogenesis in the Adult Brain Wolfgang Kelsch, Shuyin Sim, and Carlos Lois p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 131 Neurological Channelopathies Dimitri M. Kullmann p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 151 Emotion, Cognition, and Mental State Representation in Amygdala and Prefrontal Cortex C. Daniel Salzman and Stefano Fusi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 173 Category Learning in the Brain Carol A. Seger and Earl K. Miller p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 203 Molecular and Cellular Mechanisms of Learning Disabilities: A Focus on NF1 C. Shilyansky, Y.S. Lee, and A.J. Silva p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 221 Wallerian Degeneration, WldS , and Nmnat Michael P. Coleman and Marc R. Freeman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 245

Neural Mechanisms for Interacting with a World Full of Action Choices Paul Cisek and John F. Kalaska p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 269 The Role of the Human Prefrontal Cortex in Social Cognition and Moral Judgment Chad E. Forbes and Jordan Grafman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 299 Sodium Channels in Normal and Pathological Pain Sulayman D. Dib-Hajj, Theodore R. Cummins, Joel A. Black, and Stephen G. Waxman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 325
Annu. Rev. Neurosci. 2010.33:379-408. Downloaded from www.annualreviews.org by Instituto Tecnologico de Costa Rica (ITCR) on 11/02/13. For personal use only.

Mechanisms of Synapse and Dendrite Maintenance and Their Disruption in Psychiatric and Neurodegenerative Disorders Yu-Chih Lin and Anthony J. Koleske p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 349 Connecting Vascular and Nervous System Development: Angiogenesis and the Blood-Brain Barrier Stephen J. Tam and Ryan J. Watts p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 379 Motor Neuron Diversity in Development and Disease Kevin C. Kanning, Artem Kaplan, and Christopher E. Henderson p p p p p p p p p p p p p p p p p p p p p p 409 The Genomic, Biochemical, and Cellular Responses of the Retina in Inherited Photoreceptor Degenerations and Prospects for the Treatment of These Disorders Alexa N. Bramall, Alan F. Wright, Samuel G. Jacobson, and Roderick R. McInnes p p p 441 Genetics and Cell Biology of Building Specic Synaptic Connectivity Kang Shen and Peter Scheiffele p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 473 Indexes Cumulative Index of Contributing Authors, Volumes 2433 p p p p p p p p p p p p p p p p p p p p p p p p p p p 509 Cumulative Index of Chapter Titles, Volumes 2433 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 513 Errata An online log of corrections to Annual Review of Neuroscience articles may be found at http://neuro.annualreviews.org/

vi

Contents

You might also like