Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 1.
Published in final edited form as: Immunol Rev. 2012 Jul;248(1):228–239. doi: 10.1111/j.1600-065X.2012.01127.x

Inflammatory cell trafficking across the blood-brain barrier (BBB): Chemokine regulation and in vitro models

Yukio Takeshita 1,2,3, Richard M Ransohoff 1
PMCID: PMC3383666  NIHMSID: NIHMS369228  PMID: 22725965

Summary

The blood-brain barrier (BBB) is the brain-specific capillary barrier that is critical for preventing toxic substances from entering the central nervous system (CNS). In contrast to vessels of peripheral organs, the BBB limits the exchange of inflammatory cells and mediators under physiological and pathological conditions. Clarifying these limitations and the role of chemokines in regulating the BBB would provide new insights into neuroprotective strategies in neuroinflammatory diseases. Because there is a paucity of in vitro BBB models, however, some mechanistic aspects of transmigration across the BBB still remain largely unknown. In this review, we summarize current knowledge of BBB cellular components, the multi-step process of inflammatory cells crossing the BBB, functions of CNS-derived chemokines and in vitro BBB models for transmigration, with a particular focus on new and recent findings.

Keywords: in vitro BBB model, BBB components, CNS chemokine, endothelial cell line, shear stress

Cellular components of the blood-brain barrier

The blood-brain barrier (BBB) is primarily formed by microvascular endothelial cells, which are surrounded by basement membranes, pericytes, and astrocytes (Fig. 1). The endothelial basement membrane delimits the vascular aspect of the perivascular space. Astrocytic endfoot processes form the glia limitans, which, along with its own basement membrane, provide the parenchymal aspect of the perivascular space (1) (Fig. 2). This endothelial layer and glia limitans represent physical barriers to cellular entry to the central nervous system (CNS) parenchyma. Neuronal and microglial processes also contribute to the glia limitans. Interactions between endothelial cells and these surrounding cells and processes enhance BBB function and consequently result in the maintenance of proper brain homeostasis (2). More detail about each cell type is provided below.

Fig. 1. Cellular structure of the BBB.

Fig. 1

Endothelial cells have luminal tight junctions and form the capillary and the barrier. There is a basement membrane that surrounds the pericyte and astrocyte outside endothelial cells. Astrocytic endfeet are in close proximity to all of these structures.

Fig. 2. Multi-step recruitment of leukocytes into the CNS.

Fig. 2

The five steps are shown. Rolling: the binding of P-selectin and PSGL-1 in leukocytes and VCAM1 and VLA-4 in leukocytes allows the leukocyte to slow on endothelial cells; Activation: chemokines on the endothelial cells activate the rolling leukocyte; Arrest: activated leukocyte upregulates the VLA-4 and LFA-1. Binding to VCAM-1 and ICAM-1 on the endothelial cell allows the activated leukocyte attach to endothelial cells; Crawling: arrested leukocyte crawls to preferred site for migration; Migration: crawling leukocytes migrate across the endothelial cell via the paracellar or transcellar pathway. Luminal chemokines allow the crawling leukocyte to cross the endothelial cell. By abluminal chemokines leukocytes migrate to the CNS across the glia limitans. Key molecules involved in each step: PSGL-1, P-selectin glycoprotein ligand 1; VLA-4, very late antigen 4; VCAM-1, vascular cell adhesion molecule; LFA, lymphocyte function-associated antigen 1; ICAM-1, intercellular adhesion molecule-1. Adapted from (1).

Endothelial cells

Brain microvascular endothelial cells (BMVECs) directly mediate BBB function. Resting on a basement membrane, which consists mainly of collagen IV, fibronectin, laminin, and proteoglycans, BMVECs act as mediators between the blood and brain (3). They have specialized transport systems, uniform thickness with no transendothelial fenestrations, low pinocytic activity, continuous intercellular tight junctions, and high mitochondrial volume (4, 5). In addition, they have a negative luminal surface charge that repulses negatively charged compounds (5). Owing to no fenestrations and the diminished pinocytic activity, paracellular flux is limited. Uptake of essential molecules occurs through specific carrier and transport systems (6, 7). In addition, because of the presence of continuous tight junctions and adherens junctions, the paracellular space between adjacent lateral endothelial membranes is almost completely sealed (711). As they have a greater number and volume of mitochondria compared with endothelia in other organs, brain endothelial cells can provide energy and regulate the selective transport and metabolism of substances from blood to brain as well as from the parenchyma back to the systemic circulation (12).

Astrocytes

Astrocytes are important components of the BBB. Astrocytic endfeet ensheathe 99% of the surface of brain microvessels from which their endfoot processes are separated only by a thin basal membrane (13). Astrocytes are a source of important regulatory factors such as transforming growth factor-β (TGF-β) (14), glial-derived neurotropic factor (GDNF) (15), and the fibroblast growth factor (FGF) (16), and they can provide these secreted factors to endothelial cells (17). The current predominant view is that astrocytes regulate various aspects of BBB physiology with secreted factors and influence particular BBB features such as permeability, leading to tight junction formation and expression in endothelial cells (18).

Pericytes

Pericytes are important cellular constituents of capillaries and post-capillary venules. They share the same basement membrane with the endothelial cell (19) and cover 22–32% of the capillaries in CNS (20). The extent of pericyte coverage of BBB vessels is highest among the varied types of vessels (21). Pericytes regulate many neurovascular functions such as angiogenesis, BBB formation in embryogenesis, maintenance, vascular stability, regulation of capillary blood flow, and clearance of toxic cellular products (22). Pericytes can control the expression of tight junction molecules in endothelial cells by secreting factors such as TGF-β, (14) and angiopoietin (23). TGF-β enhances BBB function by inhibiting the migration of leukocytes and the proliferation of endothelial cells. The release of angiopoietin can induce remodeling and stabilization of capillaries. Platelet-derived growth factor-β (PDGFβ), secreted by endothelial cells, is an essential factor for recruitment and maintenance of pericytes on vessels and vascular maturation (24, 25).

Other cell types

The interaction between endothelial cells and neurons plays an essential role in the neurovascular network. Neurons can regulate BBB function by expressing BBB-related enzymes (26). Microglial endfoot processes, found in the perivascular glia limitans, are hypothesized to influence BBB properties. However, their contribution to BBB function remains unknown.

Multi-step process of leukocytes crossing into the CNS

Intrusion of leukocytes into CNS

The CNS exhibits strictly controlled inflammatory reactions, in part because the BBB and other vascular-tissue barriers limit the exchange of inflammatory cells and mediators. There are several routes that leukocytes can use to enter into CNS: migration from the microvessels into parenchymal perivascular space, migration via the choroid plexus into the cerebrospinal fluid, and migration through post-capillary venules at the pial surface into subarachnoid and Virchow-Robin perivascular spaces (2729). A fourth route has also been suggested that involves migration from subependymal vessels via the ependyma into the ventricles (30). These routes involve crossing the BBB, the blood–cerebrospinal fluid (CSF) barrier and the blood–spinal cord (BSC) barrier (27, 31).

Multi-step process of crossing into the CNS

Transendothelial leukocyte migration through the BBB is a multi-step process characterized by a series of sequential and tightly controlled steps that follow the paradigm of leukocyte extravasation across all vascular beds (1, 3234) (Fig. 2). The steps are: (i) rolling: weak adhesion of leukocytes to endothelial cells mainly through interactions between selectins and their carbohydrate counter-receptors; (ii) activation: leukocyte activation through chemokine stimulation of G-protein-linked receptors, resulting in functional activation of adhesion molecules along their surface; (iii) arrest: leukocyte attachment to endothelial cells through interactions between integrins associated with leukocytes and cell adhesion molecules (CAMs) on endothelial cells; (iv) crawling: leukocytes seeking preferred sites of transmigration across the endothelium; (v) transmigration: migration of leukocytes across CNS endothelia into the perivascular space and progression across the glia limitans into the brain parenchyma, a process driven in part by chemokine–chemokine receptor interactions. By interacting pairs of selectins and their ligands, integrins and CAMs, and chemokines and chemokine receptors, brain-specific processes are determined. Each of these steps and interacting pairs is described in more detail below.

Rolling

This multi-step process starts with a short and initial transient contact of the circulating leukocytes with the endothelial cell through E- and P-selectin and carbohydrate adducts on their leukocyte ligand P-selectin glycoprotein 1 (PSGL1) (35). Very late antigen-4 (VLA-4) can also support rolling. The interactions between selectins and their ligands are of low-affinity and leukocytes roll along the vascular wall with gradually reduced velocity. Recent studies have shown that despite the blockade or absence of P-selectin, immune-reactions induced in mouse models of experimental autoimmune encephalomyelitis (EAE) are indistinguishable from wildtype EAE (36, 37). This observation suggests that P-selectin is not required for leukocytes to migrate across the CNS parenchymal vessels. On the other hand, P-selectin is stored in the Weibel-Palade bodies of endothelial cells of meningeal and the fenestrated choroid plexus capillaries (38, 39). P-selectin is believed to be important for leukocyte recruitment across meningeal and choroid plexus vessels (40).

Activation

Rolling along the vascular wall slows circulating lymphocytes and permits factors such as chemokines, immobilized on the endothelial cell surface, to activate integrins on leukocytes (37, 41, 42). Chemokine receptors such as CXCR4 on rolling leukocytes interact with chemokines such as CXCL12 on endothelial cells. Chemokine receptors enhance a G-protein intracellular signal, which induces conformational changes of leukocyte integrins. Chemokines activate several signaling pathways (PI3K, PLC, RAS- and RHO-family GTPase, and MAPK), leading to opened integrin conformation (4345). As a result, adhesion molecules such as LFA-1 and VLA-4 are activated on the leukocyte surface. Integrin activation leads to enhanced avidity and affinity of the leukocyte integrin for its endothelial ligands, specifically VCAM-1 or fibronectin CS1 epitope (46, 47) and intercellular adhesion molecule-1 (ICAM-1).

Arrest

During arrest, adhesion molecules in leukocytes (VLA-4, LFA-1, and Mac-1) and endothelial receptors such as ICAM-1 and VCAM-1 play important roles (32). ICAM-1 and VCAM-1 are the major ligands for leukocyte integrins to attach to the endothelial cell against shear flow. Under normal conditions, ICAM-1 is detected on a small number of CNS microvessels and strongly upregulated by inflammatory stimuli (48). In contrast, VCAM-1 upregulation on human CNS microvessels is still matter of debate (4951). Binding of these integrins to their endothelial ligands, such as VLA-4/VCAM-1 and LFA-1, Mac-1/ICAM-1, generates cytoplasmic signaling cascades in both leukocytes and endothelial cells. As a result, leukocytes arrest on the endothelial cells.

Crawling

After leukocytes arrest, they crawl via tightly regulated integrin/CAM interactions (LFA-1/ICAM-1, VLA-4/VCAM-1) (52). These interactions initiate essential signaling within the endothelial cells and promote indentification of optimal sites for transmigration (53). Leukocytes crawl inside blood vessels in a MAC1- and ICAM1-dependent manner (52, 54). Recently an immobilized intravascular gradient of the chemokine CXCL-1 was shown to guide crawling neutrophils to transmigration sites (55). It is unknown if CNS-derived chemokines play a similar role by directing leukocytes crawling against the direction of blood flow.

Transmigration

The last stage in the multi-step process is transmigration. It is not clear whether the leukocytes cross the endothelial cell through tight junctions, via a large pore or vacuole in the endothelial cell, or through some other site (56). Until recently, leukocyte migration across the endothelial cell was thought to occur through the paracellular pathway only but leukocyte migration through the transcellular route occurs in the CNS, and in various inflammatory conditions (57, 58), and in vitro models (5961). Transmigration of leukocytes appears to be regulated by CAMs (ICAM-1, VCAM-1) and chemokine signaling processes (32). If crawling is inhibited, transmigration is delayed and occurs preferentially through the transcellular pathway as opposed to the paracellular pathway (53). While in peripheral tissues migrated cells directly enter the tissue parenchyma, in the CNS, migrated cells can only access perivascular spaces. To access the CNS parenchyma, they need to reach beyond the glia limitans, which is unique to the architecture of the BBB. Currently, there are no suitable in vitro BBB models to analyze transmigration of cells, therefore it remains incompletely understood which molecules including chemokines and chemokine receptors are critical for this process.

Chemokines and chemokine receptors

Chemokines

Chemokines play critical roles in the initial inflammatory recruitment of leukocytes. In addition to leukocyte chemotaxis, chemokines are involved in neuronal positioning during development, modulating synaptic transmission, regulating cell adhesion, phagocytosis, cytokine secretion, matrix metalloproteinase release, T-cell differentiation and activation, apoptosis, and angiogenesis (6265).

Chemokines are a group of small (8–14 kDa) structurally related molecules released by a variety of cell types. Approximately 50 human chemokine genes have been identified to date (Table 1). In spite of a variable amino acid sequence, all chemokines share a characteristic tertiary structure called the ‘chemokine fold’(66). Chemokines are divided into four subfamilies according to the configuration of two positionally conserved cysteine residues near the N-terminus. These include the CC subfamily (CCL1-CCL28), CXC subfamily (CXCL1-CXCL16), C subfamily (XCL1-XCL2), and CX3C subfamily (CX3CL1) and their nomenclature has been reviewed (66, 67).

Table 1.

Chemokines and their related receptors (67, 72,154157).

Subfamily
 Subgroup
  Chemokine name Alternative name Chemokine Receptor
CXC family
 ELR motif(+)
  CXCL1 Gro-alpha, MGSA, N51/KC, MIP-2 CXCR2
  CXCL2 Gro-beta, MIP-2 alpha CXCR2
  CXCL3 Gro-gamma, MIP-2 beta CXCR2
  CXCL5 ENA-78 CXCR2
  CXCL6 GCP-2 CXCR1, CXCR2
  CXCL7 beta-TG, CTAP-III, NAP-2 CXCR2
  CXCL8 IL-8 CXCR1, CXCR2
  CXCL15 Lungkine Unknown
 ELR motif(−)
  CXCL4 Platelet factor 4(PF4) Unknown
  CXCL9 MIG CXCR3
  CXCL10 IP10, CRG-2 CXCR3
  CXCL11 I-TAC, beta-R1, IP9, H174 CXCR3, CXCR7
  CXCL12 SDF-1 alpha, SDF1 beta, PBSF CXCR4, CXCR7
  CXCL13 BCA-1, BLC CXCR5
  CXCL14 BRAK, bolekine Unknown
  CXCL16 SR-PSOX CXCR6
CC family
  CCL1 I-309 CCR8
  CCL2 MCP-1 CCR2
  CCL3 MIP-1, LD78 CCR1, CCR5
  CCL4 MIP-1, Act-2 CCR5
  CCL5 RANTES CCR1, CCR3, CCR5
  CCL6 mC10 CCR1
  CCL7 MCP-3, FIC, MARC CCR1, CCR2, CCR3, CCR5
  CCL8 MCP-2 CCR1, CCR2, CCR3, CCR5
  CCL9/10 MIP-1gammma CCR1
  CCL11 Eotaxin CCR3, CCR5
  CCL12 MCP-5 CCR2
  CCL13 MCP-4, CK10 CCR1, CCR2, CCR3, CCR5
  CCL14 HCC, CK1 CCR1
  CCL15 HCC-2, MIP-5, MIP-1 CCR1, CCR3
  CCL16 HCC-4, CK12 CCR1
  CCL17 TARC CCR4
  CCL18 DC-CK1, PARC, MIP-4, CK7 Unknown
  CCL19 MIP-3 CCR7, CCR11
  CCL20 MIP-3, LARC, Exodus-1, CK4 CCR6
  CCL21 SLC, 6Ckine, Exodus-2, TCA4 CCR7, CCR11, CXCR3,
  CCL22 MDC CCR4
  CCL23 MPIF-1, CK8, MIP-3 CCR1,CCR12
  CCL24 MPIF-2, CK6, Eotaxin-2 CCR3
  CCL25 TECK, CK15 CCR9, CCR11
  CCL26 Eotaxin-3, MIP-4 CCR3, CCR10
  CCL27 CTAK, Eskine CCR10
  CCL28 skinkine, MEC CCR3, CCR10
C family
  XCL1 Lymphotactin alpha, SCM-1 alpha,ATAC alpha XCR1
  XCL2 Lymphotactin beta, SCM-1 beta, ATAC beta XCR1
CX3C family
  CX3CL1 Fractalkine CX3CR1

CC chemokines have a large spectrum of action and can attract monocytes, eosinophils, basophils, T lymphocytes, natural killer (NK) cells, and dendritic cells. Most CC chemokines are clustered on chromosome 17 in humans.

The CXC chemokines are distinguished by the presence or absence of a specific amino acid sequence, called the ELR-motif (glutamic acid-leucine-arginine) located near the N-terminus. The ELR+ CXC chemokines bind the neutrophil receptors CXCR2 and some also bind CXCR1. On the other hand, the ELR- CXC chemokines are inactive towards neutrophils but are potent chemoattractants for other leukocytes appropriate receptors (68).

The C chemokines, which comprise XCL1 and XCL2, are distinguished from the other chemokine subfamilies by the presence of only two of the four conserved cysteine residues (69). C chemokines chemoattract lymphocytes but not neutrophils or monocytes.

The CX3C chemokine is CX3CL1, which is characterized by the presence of three amino acids between the first two cysteine residues as well as transmembrane and mucin-like domains in C-terminal sequence. CX3CL1 can be soluble or membrane-bound (70) and acts as an adhesion molecule or a chemoattractant for T lymphocytes, NK cells, and mononuclear phagocytes (71).

Chemokine receptors

Chemokines exert their biological functions by binding to seven transmembrane-domain receptors on target cells. Chemokine receptors are classified according to the ligand family to which they respond (Table 2). The 19 known receptors often bind multiple chemokines in a subclass-restricted manner although some (such as CCR1) are highly promiscuous, while others (such as CCR8) respond only to a single unique ligand. Chemokine receptors are rhodopsin-like G protein-coupled receptors, with an acidic N-terminal extracellular domain and serine/threonine-rich intracellular C-terminal domain (72). Some chemokine receptors are widely expressed throughout the entire body, whereas others are expressed in certain specific cells or tissues or in specific activation or differentiation states of the receptor-bearing cell (73).

Table 2.

Chemokines receptors and their related chemokines (68, 71,157160).

Receptor Chemokine Ligands
CXC family
 CXCR1 CXCL6, CXCL8
 CXCR2 CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, CXCL8
 CXCR3 CXCL9, CXCL10, CXCL11
 CXCR4 CXCL12
 CXCR5 CXCL13
 CXCR6 CXCL16
 CXCR7 CXCL11, CXCL12
CC family
 CCR1 CCL2, CCL3, CCL5, CCL6, CCL7, CCL8, CCL9/10, CCL13, CCL14, CCL15, CCL16, CCL23
 CCR2 CCL2, CCL7, CCL8, CCL12, CCL13
 CCR3 CCL5, CCL7, CCL8, CCL11, CCL13, CCL15, CCL24, CCL26, CCL28
 CCR4 CCL17, CCL22
 CCR5 CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL15
 CCR6 CCL20
 CCR7 CCL19, CCL21
 CCR8 CCL1
 CCR9 CCL25
 CCR10 CCL27, CCL28
 CCR11 CCL19, CCL21, CCL25
 CCR12 CCL23
C family
 XCR1 XCL1, XCL2
CX3C family
 CX3CR1 CX3CL1

CNS chemokines and receptors

The expression of chemokines and their receptors in the CNS has been described by several authors through immunohistochemistry. It has been difficult to produce specific and sensitive antibodies for chemokines and receptors. Unfortunately, many preliminary reports could not be confirmed by critical studies using wildtype and gene-deficient mice (74). As a result, compared to peripheral tissues, chemokine functions in the CNS are less known. Chemokines and receptors that are constitutively expressed or developmentally regulated in the CNS include CXCL12-CXCR4/CXCR7, CXCL1-CXCR2, and CX3CL1-CX3CR1. CXCL12-CXCR4, which are selectively expressed in the developing and adult brain, control the migration and survival of neural precursors and stimulate astrocyte proliferation. The functions of CXCL12-CXCR7 still remain incompletely understood (75). CXCL1-CXCR2 are also implicated in the migration and proliferation of oligodendrocyte progenitors (76, 77). CX3CL1-CX3CR1, which are constitutively expressed in CNS modify inflammatory reactions of microglia and are required for recruitment of NK cells (7882).

Chemokine control of cell migration

Chemokine signaling results in molecular and functional changes in leukocytes. Chemokines and their receptors are involved in multiple steps during leukocyte transendothelial migration (Fig. 2). Chemokines presented on luminal endothelial surfaces can trigger integrin activation. As representative arrest chemokines, CXCL12, CCL11, and CCL21 can trigger integrin-dependent adhesion of leukocytes, preceding crawling towards interendothelial junctions (1, 8388).

There are other important molecules in this step, namely Duffy antigen receptor for chemokines (DARC) and D6. DARC can bind multiple chemokines from CXC and CC subfamilies, although their binding does not induce G protein-coupled cellular responses (89, 90). DARC is expressed on endothelium of capillaries and post-capillary venules, transferring chemokines across the endothelium to the lumen where the chemokine can be bound to glycosaminoglycans (GAGs) or ‘presented’ by Duffy (91,92). As a result, chemokines can be immobilized at high local concentrations on endothelial cells in the flowing blood. D6 another chemokine receptor like molecule that lacks G-protein coupling can bind to at least 12 CC chemokines (93) and is expressed on lymphatic endothelial cells, where it controls tissue concentration of CC chemokines by internalizing and degrading its ligands (94, 95).

In vitro dynamic model of BBB

After leukocyte arrest on the vascular lumen, signaling from chemokines on the abluminal aspect of the endothelium may initiate leukocyte transmigration. Using a modified Boyden chamber and human umbilical vein endothelial cells (HUVECs) under physiological flow conditions, it was shown that CXCL12 on the luminal side induces two steps in transendothelial migration for T-lymphocytes: arrest and crawling on activated HUVEC layers under shear forces. These actions of CXCL12 enhance final transmigration to abluminal CCL5, a weak subendothelial chemokine stimulus (96). This research set the stage for development of a new generation of in vitro BBB models.

Leukocyte migration across the BBB has been shown in many neurological disorders such as multiple sclerosis and stroke (97, 98), but the precise functions of chemokines in mediating leukocyte-endothelial interactions at the BBB remain incompletely understood. Models of the human BBB are being developed to address these issues. The existence of a large number of different in vitro models suggest that there is no one perfect model system and that certain models can be advantageous in specific situations. To closely mimic in vivo conditions, in vitro BBB models should have four important properties. Initially, cells for in vitro experiments should be isolated from human sources and their physiological and morphological properties should remain consistent and BBB-like. Secondly, endothelial cells should be co-cultured with other BBB components. Thirdly, the model should incorporate shear forces. Finally, the model should allow the transendothelial migration of inflammatory cells that can be recovered for further analysis. Further, the model should permit addition of chemokines.

Cell lines for BBB experiments

Primary cultures of brain microvascular endothelial cells (BMECs) represent the closest possible approximation to the in vivo BBB (99). The most widely used primary BMECs originate from rat, mouse, pig, and cow (100). The use of human BMECs is rare and limited (101, 102) due to the restricted availability of human brain tissue as well as the high cost and special skills necessary for isolation and culture of primary human BMECs. Unfortunately most primary BMECs lose their specific characteristics in culture within limited passages and rapidly cease being useful as in vitro models of the human BBB (103).

To address these issues, immortalized human BMECs were generated by expressing simian virus 40 large T antigen (SV40-LT) (104,105), human papilloma E6E7 gene (106), E1A adenovirus gene (107), or Rous sarcoma virus (108), as well as by incorporating human telomerase (109). Well-characterized human BMEC lines include human cerebral microvascular endothelial cells (hCMEC/D3) and transfected human brain microvascular endothelial cells (THBMECs) (104,109). hCMEC/D3 were established by transducing primary human brain endothelial cellswith lentiviral vectors incorporating human telomerase and SV40-LT. They have high expression of junctional proteins and have been widely used for cell signaling and drug transport studies (110119). THBMECs were isolated from human brain microvessels and immortalized by transfection with SV40-LT (104). They share characteristics of primary human BMECs including expression of tight junction-associated proteins, high transendothelial electrical resistance (TEER) (104,120), expression of factor VIII-related antigen and gamma-glutamyl transpeptidase, and uptake of 1, 1′-dioctadecyl-3, 3, 3, 3′-tetramethylindocarboxyamine perchlorate-labeled acetylated low-density lipoprotein (121). However these human BMEC lines lack contact inhibition and can lose the morphological and physiological properties of their in vivo siblings because of their immortalization particularly at high passage number or super-confluence. Under those conditions, they can present transudative intercellular junctions and lack paracellular barrier properties, which limit their effective use as an in vitro BBB model (122). Moreover, complex karyotype changes were recently reported in immortalized BMECs, rendering important the genetic testing of cell lines before their application to in vitro studies (123). As a general statement, there are few cell lines that are appropriate for in vitro BBB experiments.

Anew conditionally immortalized human BMEC cell line was established recently using a temperature-sensitive SV40-LT in order to improve BBB-like differentiated characteristics of these immortalized cell lines (124). At 33°C, SV40-T antigen binds and inhibits p53 and Rb, which are strong tumor suppressors, leading to continuous cell proliferation. At 37°C, SV40-LT is inactivated, and the cells exhibit growth arrest and differentiate into endothelial cells. These conditionally immortalized cells express occludin and claudin-5 at intercellular boundaries as well as influx and efflux transporters. At 37°C, conditional immortalized human BMEC cells retain the physiological and morphological properties of human BMECs and may represent a usefull cellular model for in vitro experiments.

In vitro BBB model for co-culture experiments

Co-culture systems, incorporating communication between endothelial cells and other BBB components, provide a closer reproduction of in vivo conditions. A significant step towards the understanding of co-culture models was the discovery that glial cells enhanced BBB properties (125). After hollow fibers with transmural microperforations were generated, many in vitro co-culture models incorporating brain endothelial cells and glial cells were developed.

Astrocyte endfeet are the cell components in closest proximity to brain capillary endothelial cells (126130). Most co-culture BBB models focused on reconstructing the brain microenvironment by incorporating astrocyte co-cultures, or astrocyte-conditioned medium, to further induce BMECs (126, 131135). It is now possible to evaluate endothelial cells in the presence of other types of cells, such as pericytes (136, 137), neurons (138), and microglia (139). Moreover, studies have been conducted with triple cultures of BMVECs, astrocytes, and pericytes (103,140), as well as with BMVECs, astrocytes and neurons (141), all extracted from rodent brain. The establishment of human multi- culture systems using BMECs and other BBB components is challenging because of the limited availability of human cell lines and the complexity of these multi- culture systems (142145).

Dynamic model of BBB for shear stress

There is increasing evidence that shear stress affects endothelial-leukocyte interactions in a complex and subtle fashion (146). This understanding led to the development of dynamic in vitro models. Among the first dynamic models were co-cultures of bovine aortic endothelial cells and glial cells (135, 147). Recently a dynamic model allowing pulsatile flow and using hCMEC/D3 cell line and astrocytes was developed (134, 148). The model showed much higher TEER than static models. Now that it is clear that shear stress allows in vitro endothelial cells to incorporate many physiological, anatomical, and biochemical BBB characteristics, including leukocyte transmigration and drug-resistant properties (134, 135, 149, 150), flow-based models are beginning to be applied for in vitro BBB studies.

Dynamic model of BBB for transmigration in response to chemokines Pioneering flow-based models showed convincingly that shear forces in the presence of chemokines control the processes of leukocyte transmigration, including arrest and crawling (83, 96, 151, 152). Recently, due to the construction of more physiological shear stress systems and the development of hollow fiber technology, some attractive artificial BBB models for migration have been developed (149, 153). For example, Man et al. (149) constructed a model that allows the transendothelial migration of inflammatory cells with the addition of chemokines. This model demonstrated that monocytes selectively adhered to BBB endothelium in response to CXCL12 and facilitated lymphocyte migration across BBB. This model provides a three-dimensional, controllable and physiologically relevant environment where vascular endothelial cells can be exposed to physiological levels of flow and chemokines, and might be useful for future transmigration experiments.

Acknowledgments

Y.T. thanks Birgit Obermeier and Anne Cotleur and Chris Nelson for reading and commenting on this manuscript and Miyuki Takeshita for help with the figures. Our research has been supported by the National Institutes of Health (K24NS51400, R21NS78420, and P50NS38667, project 1 to RMR) and Grant-in-Aid fellowship from the Japan Society for Promotion of Science (JSPS).

Footnotes

The authors have no conflicts of interest to declare.

References

  • 1.Man S, Ubogu EE, Ransohoff RM. Inflammatory cell migration into the central nervous system: a few new twists on an old tale. Brain Pathol. 2007;17:243–250. doi: 10.1111/j.1750-3639.2007.00067.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Choi YK, Kim K-W. Blood-neural barrier: its diversity and coordinated cell-to-cell communication. BMB Rep. 2008;41:345–352. doi: 10.5483/bmbrep.2008.41.5.345. [DOI] [PubMed] [Google Scholar]
  • 3.AR, Shusta EV. A genomic comparison of in vivo and in vitrobrain microvascular endothelial cells. J Cerebr Blood F Met. 2008;28:135–148. doi: 10.1038/sj.jcbfm.9600518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chaudhuri JD. Blood brain barrier and infection. Med Sci Mon Int Med J Exp Clin Res. 2000;6:1213–1222. [PubMed] [Google Scholar]
  • 5.de Boer AG, Gaillard PJ. Blood-brain barrier dysfunction and recovery. J Neural Transm. 2006;113:455–462. doi: 10.1007/s00702-005-0375-4. [DOI] [PubMed] [Google Scholar]
  • 6.Gloor SM, Wachtel M, Bolliger MF, Ishihara H, Landmann R, Frei K. Molecular and cellular permeability control at the blood-brain barrier. Brain Res Rev. 2001;36:258–264. doi: 10.1016/s0165-0173(01)00102-3. [DOI] [PubMed] [Google Scholar]
  • 7.Kniesel U, Wolburg H. Tight junctions of the blood-brain barrier. Cell Mol Neurobiol. 2000;20:57–76. doi: 10.1023/A:1006995910836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schulze C, Firth JA. Immunohistochemical localization of adherens junction components in blood-brain barrier microvessels of the rat. J Cell Sci. 1993;104:773–782. doi: 10.1242/jcs.104.3.773. [DOI] [PubMed] [Google Scholar]
  • 9.Wolburg H, Lippoldt A. Tight junctions of the blood-brain barrier: development, composition and regulation. Vasc Pharmacol. 2002;38:323–337. doi: 10.1016/s1537-1891(02)00200-8. [DOI] [PubMed] [Google Scholar]
  • 10.Vorbrodt AW, Dobrogowska DH. M olecular anatomy of intercellular junctions in brain endothelial and epithelial barriers: electron microscopist’s view. Res Dev Disabil. 2003;42:221–242. doi: 10.1016/s0165-0173(03)00177-2. [DOI] [PubMed] [Google Scholar]
  • 11.Harhaj NS, Antonetti DA. Regulation of tight junctions and loss of barrier function in pathophysiology. Int J Biochem Cell Biol. 2004;36:1206–1237. doi: 10.1016/j.biocel.2003.08.007. [DOI] [PubMed] [Google Scholar]
  • 12.Zheng W, Aschner M, Ghersi-Egea JF. Brain barrier systems: a new frontier in metal neurotoxicological research. Toxicol Appl Pharmacol. 2003;192:1–11. doi: 10.1016/s0041-008x(03)00251-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hawkins BT, Davis TP. The Blood-Brain Barrier / Neurovascular Unit in Health and Disease. Pharmacol Rev. 2005;57:173–185. doi: 10.1124/pr.57.2.4. [DOI] [PubMed] [Google Scholar]
  • 14.Dohgu S, et al. Brain pericytes contribute to the induction and up-regulation of blood-brain barrier functions through transforming growth factor-beta production. Brain Res. 2005;1038:208–215. doi: 10.1016/j.brainres.2005.01.027. [DOI] [PubMed] [Google Scholar]
  • 15.Igarashi Y, et al. Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood-brain barrier. Biochem Biophys Res Comm. 1999;261:108–112. doi: 10.1006/bbrc.1999.0992. [DOI] [PubMed] [Google Scholar]
  • 16.Reuss B, Dono R, Unsicker K. Functions of fibroblast growth factor (FGF)-2 and FGF-5 in astroglial differentiation and blood-brain barrier permeability: evidence from mouse mutants. J Neurosci. 2003;23:6404–6412. doi: 10.1523/JNEUROSCI.23-16-06404.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mi H, Haeberle H, Barres BA. Induction of astrocyte differentiation by endothelial cells. J Neurosci. 2001;21:1538–1547. doi: 10.1523/JNEUROSCI.21-05-01538.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Alvarez JI, Cayrol R, Prat A. Disruption of central nervous system barriers in multiple sclerosis. Biochim Biophys Acta. 2011;1812:252–264. doi: 10.1016/j.bbadis.2010.06.017. [DOI] [PubMed] [Google Scholar]
  • 19.Bagley RG, Weber W, Rouleau C, Teicher BA. Pericytes and endothelial precursor cells: cellular interactions and contributions to malignancy. Canc Res. 2005;65:9741–9750. doi: 10.1158/0008-5472.CAN-04-4337. [DOI] [PubMed] [Google Scholar]
  • 20.Dore-Duffy P. Pericytes: pluripotent cells of the blood brain barrier. Curr Pharmaceut Des. 2008;14:1581–1593. doi: 10.2174/138161208784705469. [DOI] [PubMed] [Google Scholar]
  • 21.Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circ Res. 2005;97:512–523. doi: 10.1161/01.RES.0000182903.16652.d7. [DOI] [PubMed] [Google Scholar]
  • 22.Winkler EA, Bell RD, Zlokovic BV. Central nervous system pericytes in health and disease. Nat Neurosci. 2011;14:1398–1405. doi: 10.1038/nn.2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hori S, Ohtsuki S, Hosoya K, Nakashima E, Terasaki T. A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro. J Neurochem. 2004;89:503–513. doi: 10.1111/j.1471-4159.2004.02343.x. [DOI] [PubMed] [Google Scholar]
  • 24.Lindahl P, Johansson BR, Levéen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science. 1997;277:242–245. doi: 10.1126/science.277.5323.242. [DOI] [PubMed] [Google Scholar]
  • 25.Leveen P, Pekny M, Gebre-Medhin S, Swolin B, Larsson E, Betsholtz C. Mice deficient for PDGF B show renal, cardiovascular, and hematological abnormalities. Gene Dev. 1994;8:1875–1887. doi: 10.1101/gad.8.16.1875. [DOI] [PubMed] [Google Scholar]
  • 26.Tontsch U, Bauer HC. Glial cells and neurons induce blood-brain barrier related enzymes in cultured cerebral endothelial cells. Brain Res. 1991;539:247–253. doi: 10.1016/0006-8993(91)91628-e. [DOI] [PubMed] [Google Scholar]
  • 27.Engelhardt B, Ransohoff RM. The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms. Trends Immunol. 2005;26:485–495. doi: 10.1016/j.it.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 28.Engelhardt B. Regulation of immune cell entry into the central nervous system. Results Probl Cell Differ. 2006;43:259–280. doi: 10.1007/400_020. [DOI] [PubMed] [Google Scholar]
  • 29.Ransohoff RM, Kivisäkk P, Kidd G. Three or more routes for leukocyte migration into the central nervous system. Nat Rev Immunol. 2003;3:569–581. doi: 10.1038/nri1130. [DOI] [PubMed] [Google Scholar]
  • 30.Alvarez JI, Teale JM. Evidence for differential changes of junctional complex proteins in murine neurocysticercosis. doi: 10.1016/j.brainres.2007.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ubogu EE, Cossoy MB, Ransohoff RM. The expression and function of chemokines involved in CNS inflammation. Trends Pharmacol Sci. 2006;27:48–55. doi: 10.1016/j.tips.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 32.Greenwood J, Heasman SJ, Alvarez JI, Prat a, Lyck R, Engelhardt B. Review: leucocyte-endothelial cell crosstalk at the blood-brain barrier: a prerequisite for successful immune cell entry to the brain. Neuropathol Appl Neurobiol. 2011;37:24–39. doi: 10.1111/j.1365-2990.2010.01140.x. [DOI] [PubMed] [Google Scholar]
  • 33.Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–689. doi: 10.1038/nri2156. [DOI] [PubMed] [Google Scholar]
  • 34.Engelhardt B. T cell migration into the central nervous system during health and disease: Different molecular keys allow access to different central nervous system compartments. Society. 2010;1:79–93. [Google Scholar]
  • 35.Kerfoot SM, Kubes P. Overlapping roles of P-selectin and alpha 4 integrin to recruit leukocytes to the central nervous system in experimental autoimmune encephalomyelitis. J Immunol. 2002;169:1000–1006. doi: 10.4049/jimmunol.169.2.1000. [DOI] [PubMed] [Google Scholar]
  • 36.Kerfoot SM, Norman MU, Lapointe BM, Bonder CS, Zbytnuik L, Kubes P. Treatment of Experimental Autoimmune Encephalomyelitis. J Immunol. 2006;176:6225–6234. doi: 10.4049/jimmunol.176.10.6225. [DOI] [PubMed] [Google Scholar]
  • 37.Engelhardt B. The blood-central nervous system barriers actively control immune cell entry into the central nervous system. Curr Pharmaceut Des. 2008;14:1555–1565. doi: 10.2174/138161208784705432. [DOI] [PubMed] [Google Scholar]
  • 38.Mayadas TN, Johnson RC, Rayburn H, Hynes RO, Wagner DD. Leukocyte rolling and extravasation are severely compromised in P selectin-deficient mice. Cell. 1993;13:541–554. doi: 10.1016/0092-8674(93)80055-j. [DOI] [PubMed] [Google Scholar]
  • 39.Kivisäkk P, et al. Human cerebrospinal fluid central memory CD4+ T cells: evidence for trafficking through chroid plexus and meninges via P-selectin. Proc Natl Acad Sci USA. 2003;100:8389–8394. doi: 10.1073/pnas.1433000100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Carrithers MD, Visintin I, Kang SJ, Janeway CA., Jr Differential adhesion molecule requirements for immune surveillance and inflammatory recruitment. Brain. 2000;123:1092–1101. doi: 10.1093/brain/123.6.1092. [DOI] [PubMed] [Google Scholar]
  • 41.Johnston B, Butcher EC. Chemokines in rapid leukocyte adhesion triggering and migration. Semin Immunol. 2002;14:83–92. doi: 10.1006/smim.2001.0345. [DOI] [PubMed] [Google Scholar]
  • 42.Ley K. Molecular mechanisms of leukocyte recruitment in the inflammatory process. Cardiovasc Res. 1996;32:733–742. [PubMed] [Google Scholar]
  • 43.Hughes PE, Pfaff M. Integrin affinity modulation. Trends Cell Biol. 1998;8:359–364. doi: 10.1016/s0962-8924(98)01339-7. [DOI] [PubMed] [Google Scholar]
  • 44.Laudanna C, Alon R. Right on the spot. Chemokine triggering of integrin-mediated arrest of rolling leukocytes. Thromb Haemost. 2006;95:5–11. [PubMed] [Google Scholar]
  • 45.Ward SG, Marelli-Berg FM. Mechanisms of chemokine and antigen-dependent T-lymphocyte navigation. Biochem J. 2009;418:13–27. doi: 10.1042/BJ20081969. [DOI] [PubMed] [Google Scholar]
  • 46.Rosenman SJ, Shrikant P, Dubb L, Benveniste EN, Ransohoff RM. Cytokine-induced expression of vascular cell adhesion molecule-1 (VCAM-1) by astrocytes and astrocytoma cell lines. J Immunol. 1995;154:1888–1899. [PubMed] [Google Scholar]
  • 47.Man S, Tucky B, Bagheri N, Kochar R, Ransohoff RM. Alpha 4 Integrin/FN-CS1 mediated leukocyte adhesion to brain microvascular endothelial cells under flow conditions. J Neuroimmunol. 2009;29:92–99. doi: 10.1016/j.jneuroim.2009.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bö L, Peterson JW, Mørk S, Hoffman PA, Gallatin WM, Ransohoff RM, Trapp BD. Distribution of immunoglobulin superfamily members ICAM-1, -2, -3, and the beta 2 integrin LFA-1 in multiple sclerosis lesions. J Neuropathol Exp Neurol. 1996;55:1060–1072. [PubMed] [Google Scholar]
  • 49.Correale J, Villa A. The blood-brain-barrier in multiple sclerosis: functional roles and therapeutic targeting. Autoimmunity. 2007;40:148–160. doi: 10.1080/08916930601183522. [DOI] [PubMed] [Google Scholar]
  • 50.Peterson JW, Bö L, Mörk S, Chang A, Ransohoff RM, Trapp BD. VCAM-1-positive microglia target oligodendrocytes at the border of multiple sclerosis lesions. J Neuropathol Exp Neurol. 2002;61:539–546. doi: 10.1093/jnen/61.6.539. [DOI] [PubMed] [Google Scholar]
  • 51.Cannella B, Raine CS. The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol. 1995;37:424–435. doi: 10.1002/ana.410370404. [DOI] [PubMed] [Google Scholar]
  • 52.Steiner O, et al. Differential roles for endothelial ICAM-1, ICAM-2, and VCAM-1 in shear-resistant T cell arrest, polarization, and directed crawling on blood-brain barrier endothelium. J Immunol. 2010;185:4846–4855. doi: 10.4049/jimmunol.0903732. [DOI] [PubMed] [Google Scholar]
  • 53.Phillipson M, Heit B, Colarusso P, Liu L, Ballantyne CM, Kubes P. Intraluminal crawling of neutrophils to emigration sites: a molecularly distinct process from adhesion in the recruitment cascade. J Exp Med. 2006;203:2569–2575. doi: 10.1084/jem.20060925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schenkel AR, Mamdouh Z, Muller Wa. Locomotion of monocytes on endothelium is a critical step during extravasation. Nat Immunol. 2004;5:393–400. doi: 10.1038/ni1051. [DOI] [PubMed] [Google Scholar]
  • 55.Massena S, et al. A chemotactic gradient sequestered on endothelial heparan sulfate induces directional intraluminal crawling of neutrophils. Blood. 2010;116:1924–1931. doi: 10.1182/blood-2010-01-266072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Greenwood J, Howes R, Lightman S. The blood-retinal barrier in experimental autoimmune uveoretinitis. Leukocyte interactions and functional damage. Lab Investig. 1994;70:39–52. [PubMed] [Google Scholar]
  • 57.Engelhardt B, Wolburg H. Mini-review: Transendothelial migration of leukocytes: through the front door or around the side of the house? Eur J Immunol. 2004;34:2955–2963. doi: 10.1002/eji.200425327. [DOI] [PubMed] [Google Scholar]
  • 58.Feng D, Nagy JA, Pyne K, Dvorak HF, Dvorak AM. Neutrophils emigrate from venules by a transendothelial cell pathway in response to FMLP. J Exp Med. 1998;187:903–915. doi: 10.1084/jem.187.6.903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cinamon G, Shinder V, Shamri R, Alon R. Chemoattractant signals and beta 2 integrin occupancy at apical endothelial contacts combine with shear stress signals to promote transendothelial neutrophil migration. J Immunol. 2004;173:7282–7291. doi: 10.4049/jimmunol.173.12.7282. [DOI] [PubMed] [Google Scholar]
  • 60.Nieminen M, Henttinen T, Merinen M, Marttila-Ichihara F, Eriksson JE, Jalkanen S. Vimentin function in lymphocyte adhesion and transcellular migration. Nat Cell Biol. 2006;8:156–162. doi: 10.1038/ncb1355. [DOI] [PubMed] [Google Scholar]
  • 61.Millán J, Hewlett L, Glyn M, Toomre D, Clark P, Ridley AJ. Lymphocyte transcellular migration occurs through recruitment of endothelial ICAM-1 to caveola- and F-actin-rich domains. Nat Cell Biol. 2006;8:113–123. doi: 10.1038/ncb1356. [DOI] [PubMed] [Google Scholar]
  • 62.Szczuciński A, Losy J. Long-term effect of IFN-beta 1a therapy on CCL2 (MCP-1) chemokine in patients with multiple sclerosis. Folia Neuropathol. 2004;42:15–18. [PubMed] [Google Scholar]
  • 63.Biber K, Zuurman MW, Dijkstra IM, Boddeke HWGM. Chemokines in the brain: neuroimmunology and beyond. Curr Opin Pharmacol. 2002;2:63–68. doi: 10.1016/s1471-4892(01)00122-9. [DOI] [PubMed] [Google Scholar]
  • 64.Godessart N, Kunkel SL. Chemokines in autoimmune disease. Curr Opin Immunol. 2001;13:670–675. doi: 10.1016/s0952-7915(01)00277-1. [DOI] [PubMed] [Google Scholar]
  • 65.Cartier L, Hartley O, Dubois-Dauphin M, Krause K-H. Chemokine receptors in the central nervous system: role in brain inflammation and neurodegenerative diseases. Brain Res Rev. 2005;48:16–42. doi: 10.1016/j.brainresrev.2004.07.021. [DOI] [PubMed] [Google Scholar]
  • 66.Domanska UM, Kruizinga RC, den Dunnen WF, Timmer-Bosscha H, de Vries EGE, Walenkamp ME. The chemokine network, a newly discovered target in high grade gliomas. Crit Rev Oncol Hematol. 2011;79:154–163. doi: 10.1016/j.critrevonc.2010.07.006. [DOI] [PubMed] [Google Scholar]
  • 67.Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354:610–621. doi: 10.1056/NEJMra052723. [DOI] [PubMed] [Google Scholar]
  • 68.Sindern E. Role of chemokines and their receptors in the pathogenesis of multiple sclerosis. BioScience. 2004;9:457–463. doi: 10.2741/1238. [DOI] [PubMed] [Google Scholar]
  • 69.Kelner GS, et al. Lymphotactin: a cytokine that represents a new class of chemokine. Science. 1994;266:1395–1399. doi: 10.1126/science.7973732. [DOI] [PubMed] [Google Scholar]
  • 70.Bazan JF, et al. A new class of membrane-bound chemokine with a CX3C motif. Nature. 1997;385:640–644. doi: 10.1038/385640a0. [DOI] [PubMed] [Google Scholar]
  • 71.Savarin-Vuaillat C, Ransohoff RM. Chemokines and chemokine receptors in neurological disease: raise, retain, or reduce? Neurotherapeutics. 2007;4:590–601. doi: 10.1016/j.nurt.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bonecchi R, Galliera E, Borroni EM, Corsi MM, Locati M, Mantovani A. Chemokines and chemokine receptors: an overview. Front Biosci. 2009;14:540–551. doi: 10.2741/3261. [DOI] [PubMed] [Google Scholar]
  • 73.Luster AD. Chemokines--chemotactic cytokines that mediate inflammation. N Engl J Med. 1998;338:436–445. doi: 10.1056/NEJM199802123380706. [DOI] [PubMed] [Google Scholar]
  • 74.Ransohoff RM. Chemokines and chemokine receptors: standing at the crossroads of immunobiology and neurobiology. Immunity. 2009;31:711–721. doi: 10.1016/j.immuni.2009.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Dziembowska M, Tham TN, Lau P, Vitry S, Lazarini F, Dubois-Dalcq M. A role for CXCR4 signaling in survival and migration of neural and oligodendrocyte presursors. Glia. 2005;50:258–269. doi: 10.1002/glia.20170. [DOI] [PubMed] [Google Scholar]
  • 76.Padovani-Claudio DA, Liu L, Ransohoff RM, Miller RH. Alterations in the oligodendrocyte lineage, myelin, and white matter in adult mice lacking the chemokine receptor CXCR2. Glia. 2006;54:471–483. doi: 10.1002/glia.20383. [DOI] [PubMed] [Google Scholar]
  • 77.Tsai HH, et al. The chemokine receptor CXCR2 controls positioning of oligodendrocyte precursors in developing spinal cord by arresting their migration. Cell. 2002;110:373–383. doi: 10.1016/s0092-8674(02)00838-3. [DOI] [PubMed] [Google Scholar]
  • 78.Cardona AE, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:917–924. doi: 10.1038/nn1715. [DOI] [PubMed] [Google Scholar]
  • 79.Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68:19–31. doi: 10.1016/j.neuron.2010.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lee S, et al. CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol. 2010;177:2549–2562. doi: 10.2353/ajpath.2010.100265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sunnemark D, et al. CX3CL1 (fractalkine) and CX3CR1 expression in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis: kinetics and cellular origin. J Neuroinflammation. 2005;2:17. doi: 10.1186/1742-2094-2-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Huang D, et al. The neuronal chemokine CX3CL1/fractalkine selectively recruits NK cells that modify experimentalautoimmune encephalomyelitis within the central nervous system. FASEB J. 2006;20:896–905. doi: 10.1096/fj.05-5465com. [DOI] [PubMed] [Google Scholar]
  • 83.Cinamon G, Shinder V, Alon R. Shear forces promote lymphocyte migration across vascular endothelium bearing apical chemokines. Nat Immunol. 2001;2:515–522. doi: 10.1038/88710. [DOI] [PubMed] [Google Scholar]
  • 84.Weber C, Kitayama J, Springer TA. Differential regulation of beta 1 and beta 2 integrin avidity by chemoattractants in eosinophils. Proc Natl Acad Sci USA. 1996;93:10939–10944. doi: 10.1073/pnas.93.20.10939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Weber C, Alon R, Moser B, Springer TA. Sequential regulation of alpha 4 beta 1 and alpha 5 beta 1 integrin avidity by CC chemokines in monocytes: implications for transendothelial chemotaxis. J Cell Biol. 1996;134:1063–1073. doi: 10.1083/jcb.134.4.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Stein JV, et al. The CC chemokine thymus-derived chemotactic agent 4 (TCA-4, secondary lymphoid tissue chemokine, 6Ckine, exodus-2) triggers lymphocyte function-associated antigen 1-mediated arrest of rolling T lymphocytes in peripheral lymph node high endothelial venules. J Exp Med. 2000;191:61–76. doi: 10.1084/jem.191.1.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Campbell JJ, et al. The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells. Nature. 1999;400:776–780. doi: 10.1038/23495. [DOI] [PubMed] [Google Scholar]
  • 88.Campbell JJ, Hedrick J, Zlotnik A, Siani MA, Thompson DA, Butcher EC. Chemokines and the arrest of lymphocytes rolling under flow conditions. Science. 1998;279:381–384. doi: 10.1126/science.279.5349.381. [DOI] [PubMed] [Google Scholar]
  • 89.Neote K, DiGregorio D, Mak JY, Horuk R, Schall TJ. Molecular cloning, functional expression, and signaling characteristics of a C-C chemokine receptor. Cell. 1993;12:415–425. doi: 10.1016/0092-8674(93)90118-a. [DOI] [PubMed] [Google Scholar]
  • 90.Comerford I, Litchfield W, Harata-Lee Y, Nibbs RJ, McColl SR. Regulation of chemotactic networks by ‘atypical’ receptors. Bioessays. 2007;29:237–247. doi: 10.1002/bies.20537. [DOI] [PubMed] [Google Scholar]
  • 91.Lee JS, et al. Duffy antigen facilitates movement of chemokine across the endothelium in vitroand promotes neutrophil transmigration in vitro and in vivo. J immunol. 2003;15:5244–5251. doi: 10.4049/jimmunol.170.10.5244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Patterson AM, Siddall H, Chamberlain G, Gardner L, Middleton J. Expression of the duffy antigen/receptor for chemokines (DARC) by the inflamed synovial endothelium. J Pathol. 2002;197:108–116. doi: 10.1002/path.1100. [DOI] [PubMed] [Google Scholar]
  • 93.Nibbs RJ, Wylie SM, Yang J, Landau NR, Graham GJ. Cloning and characterization of a novel promiscuous human beta-chemokine receptor D6. J Biol Chem. 1997;272:32078–32083. doi: 10.1074/jbc.272.51.32078. [DOI] [PubMed] [Google Scholar]
  • 94.Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891–928. doi: 10.1146/annurev.immunol.22.012703.104543. [DOI] [PubMed] [Google Scholar]
  • 95.Rajagopalan L, Rajarathnam K. Structural basis of chemokine receptor function--a model for binding affinity and ligand selectivity. Biosci Rep. 2006;26:325–339. doi: 10.1007/s10540-006-9025-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schreiber TH, Shinder V, Cain DW, Alon R, Sackstein R. Shear flow-dependent integration of apical and subendothelial chemokines in T-cell transmigration: implications for locomotion and the multistep paradigm. Blood. 2007;109:1381–1386. doi: 10.1182/blood-2006-07-032995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Petty MA, Lo EH. Junctional complexes of the blood-brain barrier: permeability changes in neuroinflammation. Progr Neurobiol. 2002;68:311–323. doi: 10.1016/s0301-0082(02)00128-4. [DOI] [PubMed] [Google Scholar]
  • 98.Holman DW, Klein RS, Ransohoff RM. The blood-brain barrier, chemokines and multiple sclerosis. Biochim Biophys Acta. 2011;1812:220–230. doi: 10.1016/j.bbadis.2010.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Smith M, Omidi Y, Gumbleton M. Primary porcine brain microvascular endothelial cells: biochemical and functional characterisation as a model for drug transport and targeting. J Drug Target. 2007;15:253–268. doi: 10.1080/10611860701288539. [DOI] [PubMed] [Google Scholar]
  • 100.Deli MA, Ábrahám CS, Kataoka Y, Niwa M. Permeability Studies on In Vitro Blood–Brain Barrier Models: Physiology, Pathology, and Pharmacology. Cell Mol Neurobiol. 2005;25:59–127. doi: 10.1007/s10571-004-1377-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bernas MJ, et al. Establishment of primary cultures of human brain microvascular endothelial cells to provide anin vitro cellular model of the blood-brain barrier. Nat Protocol. 2010;5:1265–1272. doi: 10.1038/nprot.2010.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Siddharthan V, Kim YV, Liu S, Kim KS. Human astrocytes/astrocyte-conditioned medium and shear stress enhance the barrier properties of human brain microvascular endothelial cells. Brain Res. 2007;1147:39–50. doi: 10.1016/j.brainres.2007.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Nakagawa S, et al. A new blood-brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54:253–263. doi: 10.1016/j.neuint.2008.12.002. [DOI] [PubMed] [Google Scholar]
  • 104.Stins MF, Gilles F, Kim KS. Selective expression of adhesion molecules on human brain microvascular endothelial cells. J Neuroimmunol. 1997;76:81–90. doi: 10.1016/s0165-5728(97)00036-2. [DOI] [PubMed] [Google Scholar]
  • 105.Greenwood J, et al. SV40 large T immortalised cell lines of the rat blood-brain and blood-retinal barriers retain their phenotypic and immunological characteristics. J Neuroimmunol. 1996;71:51–63. doi: 10.1016/s0165-5728(96)00130-0. [DOI] [PubMed] [Google Scholar]
  • 106.Prudhomme JG, Sherman IW, Land KM, Moses AV, Stenglein S, Nelson JA. Studies of Plasmodium falciparum cytoadherence using immortalized human brain capillary endothelial cells. Int J Parasitol. 1996;26:647–655. doi: 10.1016/0020-7519(96)00027-6. [DOI] [PubMed] [Google Scholar]
  • 107.Roux F, et al. Regulation of gamma-glutamyl transpeptidase and alkaline phosphatase activities in immortalized rat brain microvessel endothelial cells. J Cell Physiol. 1994;159:101–113. doi: 10.1002/jcp.1041590114. [DOI] [PubMed] [Google Scholar]
  • 108.Mooradian DL, Diglio CA. Production of a transforming growth factor-beta-like growth factor by RSV-transformed rat cerebral microvascular endothelial cells. Tumour Biol. 1991;12:171–183. doi: 10.1159/000217702. [DOI] [PubMed] [Google Scholar]
  • 109.Weksler BB, et al. Blood-brain barrier-specific properties of a human adult brain endothelial cell line. Faseb J. 2005;19:1872–1874. doi: 10.1096/fj.04-3458fje. [DOI] [PubMed] [Google Scholar]
  • 110.Carl SM, et al. ABC and SLC transporter expression and pot substrate characterization across the human CMEC/D3 blood-brain barrier cell line. Mol Pharm. 2010;7:1057–1068. doi: 10.1021/mp900178j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zastre JA, et al. Up-regulation of P-glycoprotein by HIV protease inhibitors in a human brain microvessel endothelial cell line. J Neurosci Res. 2009;87:1023–1036. doi: 10.1002/jnr.21898. [DOI] [PubMed] [Google Scholar]
  • 112.Dauchy S, et al. Expression and transcriptional regulation of ABC transporters and cytochromes P450 in hCMEC/D3 human cerebral microvascular endothelial cells. Biochem Pharmacol. 2009;77:897–909. doi: 10.1016/j.bcp.2008.11.001. [DOI] [PubMed] [Google Scholar]
  • 113.Fischer S, et al. Signaling mechanism of extracellular RNA in endothelial cells. Faseb J. 2009;23:2100–2109. doi: 10.1096/fj.08-121608. [DOI] [PubMed] [Google Scholar]
  • 114.Zhong Y, Smart EJ, Weksler B, Couraud P-O, Hennig B, Toborek M. Caveolin-1 regulates human immunodeficiency virus-1 Tat-induced alterations of tight junction protein expression via modulation of the Ras signaling. J Neurosci. 2008;28:7788–7796. doi: 10.1523/JNEUROSCI.0061-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wilhelm I, et al. Hyperosmotic stress induces Axl activation and cleavage in cerebral endothelial cells. J Neurochem. 2008;107:116–126. doi: 10.1111/j.1471-4159.2008.05590.x. [DOI] [PubMed] [Google Scholar]
  • 116.Wilhelm I, et al. Regulation of cerebral endothelial cell morphology by extracellular calcium. Phys Med Biol. 2007;52:6261–6274. doi: 10.1088/0031-9155/52/20/012. [DOI] [PubMed] [Google Scholar]
  • 117.Lim JC, et al. Activation of beta-catenin signalling by GSK-3 inhibition increases p-glycoprotein expression in brain endothelial cells. J Neurochem. 2008;106:1855–1865. doi: 10.1111/j.1471-4159.2008.05537.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schreibelt G, van Horssen J, van Rossum S, Dijkstra CD, Drukarch B, de Vries HE. Therapeutic potential and biological role of endogenous antioxidant enzymes in multiple sclerosis pathology. Brain Res Rev. 2007;56:322–330. doi: 10.1016/j.brainresrev.2007.07.005. [DOI] [PubMed] [Google Scholar]
  • 119.Poller B, et al. The human brain endothelial cell line hCMEC/D3 as a human blood-brain barrier model for drug transport studies. J Neurochem. 2008;107:1358–1368. doi: 10.1111/j.1471-4159.2008.05730.x. [DOI] [PubMed] [Google Scholar]
  • 120.Callahan MK, Williams KA, Kivisäkk P, Pearce D, Stins MF, Ransohoff RM. CXCR3 marks CD4+ memory T lymphocytes that are competent to migrate across a human brain microvascular endothelial cell layer. J Neuroimmunol. 2004;153:150–157. doi: 10.1016/j.jneuroim.2004.05.004. [DOI] [PubMed] [Google Scholar]
  • 121.Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog. 2001;30:19–28. doi: 10.1006/mpat.2000.0406. [DOI] [PubMed] [Google Scholar]
  • 122.Gumbleton M, Audus KL. Progress and limitations in the use of in vitrocell cultures to serve as a permeability screen for the blood-brain barrier. J Pharmaceut Sci. 2001;90:1681–1698. doi: 10.1002/jps.1119. [DOI] [PubMed] [Google Scholar]
  • 123.Mkrtchyan H, et al. Molecular cytogenetic characterization of the human cerebral microvessel endothelial cell line hCMEC/D3. Cytogenet Genome Res. 2009;126:313–317. doi: 10.1159/000253080. [DOI] [PubMed] [Google Scholar]
  • 124.Sano Y, et al. Establishment of a new conditionally immortalized human brain microvascular endothelial cell line retaining an in vivoblood-brain barrier function. J Cell Physiol. 2010;225:519–528. doi: 10.1002/jcp.22232. [DOI] [PubMed] [Google Scholar]
  • 125.DeBault LE, Cancilla PA. Some properties of isolated endothelial cells in culture. Adv Exp Med Biol. 1980;131:69–78. doi: 10.1007/978-1-4684-3752-2_6. [DOI] [PubMed] [Google Scholar]
  • 126.Rubin LL, et al. A cell culture model of the blood-brain barrier. J Cell Biol. 1991;115:1725–1735. doi: 10.1083/jcb.115.6.1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Kacem K, Lacombe P, Seylaz J, Bonvento G. Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: a confocal microscopy study. Glia. 1998;23:1–10. [PubMed] [Google Scholar]
  • 128.Janzer RC, Raff MC. Astrocytes induce blood-brain barrier properties in endothelial cells. Nature. 1987;325:253–257. doi: 10.1038/325253a0. [DOI] [PubMed] [Google Scholar]
  • 129.Gaillard PJ, et al. Establishment and functional characterization of an in vitro model of the blood-brain barrier, comprising a co-culture of brain capillary endothelial cells and astrocytes. Eur J Pharmaceut Sci. 2001;12:215–222. doi: 10.1016/s0928-0987(00)00123-8. [DOI] [PubMed] [Google Scholar]
  • 130.Lundquist S, Renftel M, Brillault J, Fenart L, Cecchelli R, Dehouck MP. Prediction of drug transport through the blood-brain barrier in vivo: a comparison between two in vitrocell models. Pharmaceut Res. 2002;19:976–981. doi: 10.1023/a:1016462205267. [DOI] [PubMed] [Google Scholar]
  • 131.Santaguida S, Janigro D, Hossain M, Oby E, Rapp E, Cucullo L. Side by side comparison between dynamic versus static models of blood-brain barrier in vitro: a permeability study. Brain Res. 2006;1109:1–13. doi: 10.1016/j.brainres.2006.06.027. [DOI] [PubMed] [Google Scholar]
  • 132.Krizanac-Bengez L, et al. Loss of shear stress induces leukocyte-mediated cytokine release and blood-brain barrier failure in dynamic in vitro blood-brain barrier model. J Cell Physiol. 2006;206:68–77. doi: 10.1002/jcp.20429. [DOI] [PubMed] [Google Scholar]
  • 133.Parkinson FE, Friesen J, Krizanac-Bengez L, Janigro D. Use of a three-dimensional in vitro model of the rat blood-brain barrier to assay nucleoside efflux from brain. Brain Res. 2003;980:233–241. doi: 10.1016/s0006-8993(03)02980-9. [DOI] [PubMed] [Google Scholar]
  • 134.Cucullo L, et al. Immortalized human brain endothelial cells and flow-based vascular modeling: a marriage of convenience for rational neurovascular studies. J Cerebr Blood Flow Metabol. 2008;28:312–328. doi: 10.1038/sj.jcbfm.9600525. [DOI] [PubMed] [Google Scholar]
  • 135.Cucullo L, et al. A new dynamic in vitro model for the multidimensional study of astrocyte-endothelial cell interactions at the blood-brain barrier. Brain Res. 2002;951:243–254. doi: 10.1016/s0006-8993(02)03167-0. [DOI] [PubMed] [Google Scholar]
  • 136.Al Ahmad A, Gassmann M, Ogunshola OO. Maintaining blood-brain barrier integrity: pericytes perform better than astrocytes during prolonged oxygen deprivation. J Cell Physiol. 2009;218:612–622. doi: 10.1002/jcp.21638. [DOI] [PubMed] [Google Scholar]
  • 137.Kim JA, Tran ND, Li Z, Yang F, Zhou W, Fisher MJ. Brain endothelial hemostasis regulation by pericytes. J Cerebr Blood Flow Metabol. 2006;26:209–217. doi: 10.1038/sj.jcbfm.9600181. [DOI] [PubMed] [Google Scholar]
  • 138.Cestelli A, et al. Functional feature of a novel model of blood brain barrier: studies on permeation of test compounds. J Contr Release. 2001;76:139–147. doi: 10.1016/s0168-3659(01)00431-x. [DOI] [PubMed] [Google Scholar]
  • 139.Sumi N, et al. Lipopolysaccharide-activated microglia induce dysfunction of the blood-brain barrier in rat microvascular endothelial cells co-cultured with microglia. Cell Mol Neurobiol. 2010;30:247–253. doi: 10.1007/s10571-009-9446-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Nakagawa S, et al. Pericytes from brain microvessels strengthen the barrier integrity in primary cultures of rat brain endothelial cells. Cell Mol Neurobiol. 2007;27:687–694. doi: 10.1007/s10571-007-9195-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Schiera G, et al. Permeability properties of a three-cell type in vitro model of blood-brain barrier. J Cell Mol Med. 2005;9:373–379. doi: 10.1111/j.1582-4934.2005.tb00362.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Weidenfeller C, Schrot S, Zozulya A, Galla HJ. Murine brain capillary endothelial cells exhibit improved barrier properties under the influence of hydrocortisone. Brain Res. 2005;1053:162–174. doi: 10.1016/j.brainres.2005.06.049. [DOI] [PubMed] [Google Scholar]
  • 143.Perrière N, et al. Puromycin-based purification of rat brain capillaryendothelial cell cultures. Effect on the expression of blood-brain barrier-specific properties. J Neurochem. 2005;93:279–289. doi: 10.1111/j.1471-4159.2004.03020.x. [DOI] [PubMed] [Google Scholar]
  • 144.Cecchelli R, et al. Modelling of the blood-brain barrier in drug discovery and development. Nat Rev Drug Discov. 2007;6:650–661. doi: 10.1038/nrd2368. [DOI] [PubMed] [Google Scholar]
  • 145.Weidenfeller C, Svendsen CN, Shusta EV. Differentiating embryonic neural progenitor cells induce blood-brain barrier properties. J Neurochem. 2007;101:555–565. doi: 10.1111/j.1471-4159.2006.04394.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Tarbell JM. Shear stress and the endothelial transport barrier. Cardiovasc Res. 2010;87:320–330. doi: 10.1093/cvr/cvq146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Stanness KA, et al. Morphological and functional characterization of an in vitroblood-brain barrier model. Brain Res. 1997;771:329–342. doi: 10.1016/s0006-8993(97)00829-9. [DOI] [PubMed] [Google Scholar]
  • 148.Cucullo L, Marchi N, Hossain M, Janigro D. A dynamic in vitroBBB model for the studyof immune cell trafficking into the central nervous system. J Cerebr Blood Flow Metabol. 2011;31:767–777. doi: 10.1038/jcbfm.2010.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Man S, Tucky B, Cotleur A, Drazba J, Takeshita Y, Ransohoff RM. CXCL12 induced monocyte-endothelial interactions promote lymphocyte transmigration across in vitro blood-brain barrier. Sci Transl Med. 2009;119:1–10. doi: 10.1126/scitranslmed.3003197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Ghosh C, et al. Pattern of P450 expression at the human blood-brain barrier: roles of epileptic condition and laminar flow. Epilepsia. 2010;51:1408–1417. doi: 10.1111/j.1528-1167.2009.02428.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Cuvelier SL, Patel KD. Shear-dependent eosinophil transmigration on interleukin 4-stimulated endothelial cells: a role for endothelium-associated eotaxin-3. J Exp Med. 2001;194:1699–1709. doi: 10.1084/jem.194.12.1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Cinamon G, et al. Novel chemokine functions in lymphocyte migration through vascular endothelium under shear flow. J Leukoc Biol. 2001;69:860–866. [PubMed] [Google Scholar]
  • 153.Cucullo L, Hossain M, Puvenna V, Marchi N, Janigro D. The role of shear stress in Blood-Brain Barrier endothelial physiology. BMC Neurosci. 2011;12:40. doi: 10.1186/1471-2202-12-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lasagni L, et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4. J Exp Med. 2003;197:1537–1549. doi: 10.1084/jem.20021897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Belperio JA, et al. Interaction of IL-13 and C10 in the pathogenesis of bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2002;27:419–427. doi: 10.1165/rcmb.2002-0009OC. [DOI] [PubMed] [Google Scholar]
  • 156.Pan J, et al. A novel chemokine ligand for CCR10 and CCR3 expressed by epithelial cells in mucosal tissues. J Immunol. 2000;165:2943–2949. doi: 10.4049/jimmunol.165.6.2943. [DOI] [PubMed] [Google Scholar]
  • 157.Laing KJ, Secombes CJ. Chemokines. Dev Comp Immunol. 2004;28:443–460. doi: 10.1016/j.dci.2003.09.006. [DOI] [PubMed] [Google Scholar]
  • 158.Shimizu S, Brown M, Sengupta R, Penfold ME, Meucci O. CXCR7 protein expression in human adult brain and differentiated neurons. PLoS one. 2011;6:e20680. doi: 10.1371/journal.pone.0020680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Liu JX, Cao X, Tang YC, Liu Y, Tang FR. CCR7, CCR8, CCR9 and CCR10 in the mouse hippocampal CA1 area and the dentate gyrus during and after pilocarpine-induced status epilepticus. J Neurochem. 2007;100:1072–1088. doi: 10.1111/j.1471-4159.2006.04272.x. [DOI] [PubMed] [Google Scholar]
  • 160.Miao Z, et al. Proinflammatory proteases liberate a discrete high-affinity functional FPRL1(CCR12) ligand from CCL23. J Immunol. 2007;178:7395–7404. doi: 10.4049/jimmunol.178.11.7395. [DOI] [PubMed] [Google Scholar]

RESOURCES