15

Click here to load reader

Environmental influences on neural crest cell migration

Embed Size (px)

Citation preview

Page 1: Environmental influences on neural crest cell migration

Environmental Influences on Neural Crest Cell Migration

Marianne Bronner-Fraser

Developmental Biology Center, University of California, Irvine, California 9271 7

SUMMARY

Neural crest cells migrate extensively and interact with numerous tissues and extracellular matrix components during their movement. Cell marking techniques have shown that neural crest cells in the trunk of the avian embryo migrate through the anterior, but not posterior, half of each sclerotome and avoid the region around the notochord. A possible mechanism to account for this mi- gratory pattern is that neural crest cells may be inhibited from entering the posterior sclerotome and the perinoto-

chordal space. Thus, interactions with other tissue may prescribe the pattern of neural crest cell migration in the trunk. In contrast, interactions between neural crest cells and the extracellular matrix may mediate the primary interactions controlling neural crest cells migration in the head region. 0 1993 John Wiley & Sons, Inc. Keywords: neural crest cell migration, extracellular ma- trix molecules, cell marking techniques, notochord inhibi- tory effects.

INTRODUCTION

The neural crest is a migratory cell population, aris- ing within the dorsal aspect of the neural tube. After closure of the neural tube, neural crest cells emigrate from its dorsal margin, migrate along characteristic pathways, and differentiate into widely varied derivatives. The neural crest has sev- eral unique properties that make it an ideal system for studying cell migration and differentiation. First, these cells migrate extensively in a stereo- typic manner. Second, neural crest cells give rise to diverse and numerous derivatives, ranging from melanocytes and cranial cartilage to adrenal chro- maffin cells and the ganglia of the peripheral ner- vous system. Third, neural crest cells are accessible to surgical, immunological, and biochemical ma- nipulations during both initial and certain later stages in their development.

As a population, the neural crest is regionalized cells derived from different axial levels follow dis- tinct migratory pathways to their final destinations

Received September 8, 1992; accepted September 8, 1992 Journal ofNeurobiology, Vol. 24, No. 2, pp. 233-247 (1993) 0 1993 John Wiley & Sons, Inc. ccc 0022-3034/93/020233-15

and there give rise to diverse progeny (LeDouarin, 1982; Noden, 1975). In the cranial region, neural crest cells migrate subjacent to the cranial ecto- derm. Some cranial neural crest cells enter the branchial arches, where they will form many of the cartilagenous elements of the facial skeleton, whereas others contribute to the ciliary ganglion of the eye and the cranial sensory ganglia. Vagal neural crest cells migrate under the ectoderm and into gut, where they move along its anterior to pos- terior extent to populate the enteric nervous sys- tem. Trunk neural crest cells migrate in two direc- tions: ventrally through the somites and dorsally under the ectoderm. Those cells following the ven- tral route give rise to dorsal root ganglia, sympa- thetic ganglia, and adrenal chromaffin cells; those cells following the dorsal route give rise to melano- cytes of the skin (Weston, 1963; LeDouarin, 1982).

Neural crest cells migrate through or adjacent to numerous tissues including the neural tube, so- mites, and gut. Their migratory pathways often contain abundant quantities of extracellular ma- trix (ECM) molecules. In addition, neural crest cells possess receptors for ECM molecules as well as numerous cell surface molecules which may be important for cell-cell adhesion to themselves or

233

Page 2: Environmental influences on neural crest cell migration

234 Bronner-Fraser

other cell types. Thus, a variety of cell-cell and cell-substrate interactions may influence the mi- gration and localization of neural crest cells. In order to understand these interactions, one must have a detailed knowledge of the pathways fol- lowed by neural crest cells, the potential ECM li- gands available along these pathways, and the tis- sues with which neural crest cells may interact. This review summarizes recent experiments from our laboratory regarding the tissue and substrate interactions that may influence the pattern of neural crest cell migration, primarily in the trunk region, although some other populations are con- sidered as well.

ANALYSIS OF NEURAL CREST MIGRATORY PATHWAYS USING CELL MARKING TECHNIQUES

The pathways of neural crest cell migration have been studied using a variety of cell markers. The initial analysis of neural crest cell migratory path- ways in avian embryos involved transplantation of neural tubes marked with either [ 3H] thymidine (Weston, 1963) or derived from quail embryos (for review, see LeDouarin, 1982) into unlabelled chick hosts. By fixing animals at various stages after operation and looking at the distribution of labelled cells, it was possible to follow both the routes and derivatives of neural crest cells. These experiments demonstrated that neural crest cells migrate along two predominant pathways in the trunk region: either ventrally toward the dorsal aorta or dorsolaterally under the ectoderm. Such transplantation experiments have been particu- larly useful for establishing the progeny arising from neural crest cells at various axial levels.

Because transplants require some time to heal and may cause scarring, neural tube grafts have been less useful for describing the early pattern of neural crest cell migration. Recently, new tech- niques have emerged that circumvent these diffi- culties. First, monoclonal antibodies that recog- nized migrating neural crest cells and some of their derivatives have become available. NC- 1 and HNK- 1 antibodies (Tucker, Aoyama, Lipinski, Tursz, and Thiery, 1984) both recognize a carbo- hydrate epitope that is present on the surface of neural crest cells and some of their derivatives. By examining the HNK- 1 immunoreactivity in fixed sections through embryos, it was discovered that neural crest cells that move along the ventral path- way migrate in a segmental fashion, moving through the anterior, but not the posterior half of

each somitic sclerotome, and then ventrally toward the dorsal aorta (Rickmann, Fawcett, and Keynes, 1985; Bronner-Fraser, 1986a) (Fig. 1). The cells following the ventral route will form the dorsal root and sympathetic ganglion cells, as well as adrenomedullary cells. Interestingly, their meta- meric pattern of migration is later reflected in the segmental arrangement of neural crest-derived pe- ripheral ganglia (Teillet, Kalcheim, and Le Douarin, 1987; Lallier and Bronner-Fraser, 1988). Although the results obtained with antibodies are intriguing, the pattern of cell migration can be only inferred from observations of sections through fixed embryos. Furthermore, these antibodies are not entirely specific; they recognize several non- neural crest cell types (Kruse et al., 1984) and do not recognize all neural crest cells (Teillet et al., 1987).

An alternative and more direct method for ex- amining the pathways of neural crest cell migration is to inject the lipophilic dye, DiI, into the neural tube ( Serbedzija, Bronner-Fraser, and Fraser, 1989). Because the dye is hydrophobic and inter- calates into cell membranes, injection of DiI into the neural tube marks all neural tube cells includ- ing premigratorq neural crest cells within its dorsal margin. By examining embryos at various times after injection, it is possible to trace directly the routes followed by neural crest cells. Similar to the results using the HNK-1 antibody, early DiI-la- belled migrating trunk neural crest cells were found to migrate ventrally in a segmental fashion. By altering the time of injection, it also was possi- ble to establish the order in which neural crest cells contribute to their derivatives. By injecting DiI at progressively later stages of development, we la- belled only those neural crest cells that were premi- gratory at the time of injection. Later emigrating cells were found to contribute to progressively more dorsal derivatives (Fig. 2). Thus, the contri- bution of neural crest cells to their derivatives be- comes restricted in a ventral-to-dorsal order. The last cells to exit the neural tube are precursors to pigment cells, which migrate dorsolaterally under the ectoderm where they eventually form pigment cells (Serbedzija et al., 1989). An advantage of DiI- labelling of neural crest cells is that DiI can be ap- plied to a variety of axial levels and any species. For example, this approach has been used successfully to map the pathways of sacral neural crest cell mi- gration in the chick and mouse (Pomeranz and Gershon, 199 1; Serbedzija, Burgan, Fraser, and Bronner-Fraser, 199 1 ) and trunk and cranial neural crest cell migration in the mouse (Serbed- zija et al., 1990; 1992, submitted).

Page 3: Environmental influences on neural crest cell migration

Neural Crest Cell Migration 235

Figure 1 Schematic diagram illustrating the early pathways of trunk neural crest cell migra- tion. Neural crest cells emerge from the neural tube ( N T ) and proceed either ventrally (indi- cated by large curved arrow) through the somitic sclerotome (Scl) or dorso-laterally (indicated by small curved arrow) between the ectoderm (Ec) and dermomyotome (DM). The ventrally migrating cells only move through the rostra1 ( R ) half of each sclerotome and are not observed in the caudal (C) half. Neural crest cells avoid the region around the notochord (No). Ao = dorsal aorta.

The conclusion that neural crest cells migrate in an orderly pattern is in agreement with the results of Weston and Butler ( 1966), who transplanted tritiated thymidine-labelled neural tubes from the trunk region of “older” avian embryos to trunks of “younger” hosts (Weston and Butler, 1966). They found that the neural crest cells emerging from both the older and young neural tubes could give rise to the same range of neural crest derivatives. In contrast, neural crest cells emerging from “young” neural tubes transplanted to “older” embryos con- tributed only to dorsal derivatives, such as dorsal root ganglia and pigment cells. Thus, neural crest cells and/or the migratory paths available to them may change during the course of their migration and the occupation of their derivatives.

These results suggest that the time at which neural crest cells exit the neural tube may contrib-

ute to the range of derivatives available to them. This could be a result ofthe extrinsic changes occur- ring in the environment, intrinsic changes within the neural crest population, or a combination of the two. We tested whether late emigrating neural crest cells are more restricted in developmental po- tential than early migrating cells by culturing neural crest cells prepared from the neural tubes of embryos at various stages of neural crest cell migra- tion (Artinger and Bronner-Fraser, 1992). Later emigrating neural crest cells displayed a more lim- ited repertoire of cell fates than early emigrating neural crest cells. Although both populations gave rise to pigment cells and neurons, only early emi- grating neural crest cells formed adrenergic cells. These results suggest that late emigrating neural crest cells have a more restricted developmental potential than do early migrating neural crest cells.

Page 4: Environmental influences on neural crest cell migration

236 Bronner-Fraser

Figure 2 (Top) Trunk neural crest cell migration pathways and derivatives. There are two major pathways: ( A ) The dorso-lateral pathway between the somite and the ectoderm, and (B) the ventral pathway through the rostral half of each somite. Trunk neural crest give rise to (A’) pigment cells, (B’) dorsal root ganglia, ( C ’ ) sympathetic ganglia, and (D‘) cells around the dorsal aorta. (Bottom) A schematic representation of the rostral to caudal distribution of DiI in the neural crest derivatives of a single embryo injected at stage 19 and fixed at stage 2 1. (a-f) Represents levels along the rostrocaudal axis from which transverse sections were taken. Be- cause development in a single embryo occurs in a rostrocaudal sequence, different stages of neural crest migration exist within the same embryo. ( a ) At the level of the ninth somite, Dil-labelled cells were observed along the dorso-lateral pathway. (b) At the level of the 15th somite, DiI labelled cells were observed along the dorso-lateral pathway and in the dorsal root ganglia. (c ) At the level of the 22nd somite, DiI-labelled cells were seen along the dorso-lateral pathway, in the dorsal root ganglia, and in the sympathetic ganglia. (d-f) From the level of the 38th somite to the caudal end of the embryo, DiI-labelled cells were observed in all truncal neural crest derivatives. Thus, neural crest cells appear to fill their derivatives in a ventral-to- dorsal order. (From G. Serbedzija et al., Development 1062306-816, 1989.)

Page 5: Environmental influences on neural crest cell migration

Neural Crest Cell Migration 237

Thus, although the orderly pattern of migration may restrict the range of derivatives open to later emigrating neural crest cells, other factors such as time spent within the neural tube also may contrib- ute to restriction of these cells' fate.

ROLE OF SURROUNDING TISSUES IN DETERMINING THE PATTERN OF NEURAL CREST MIGRATION

Neural crest pathways are lined with a variety of tissue including the somites, ectoderm, neural tube, notochord, and dorsal aorta. During their mi- gration, neural crest cells contact a number of these tissues. Described below are experiments examin- ing the role that some of these tissues may play in establishing the pattern of neural crest cell migra- tion.

Segmental Information within the Somites

Like motor nerves (Keynes and Stern, 1984), mi- grating neural crest cells (Rickmann et al., 1985; Bronner-Fraser, 1986a; Teillet et al., 1987; Loring and Erickson, 1987) are restricted to the anterior half of each sclerotome. The conclusion that the somite contains segmental information is based on the result of a simple experiment: if the segmental plate (which gives rise to the somites) is rotated 180" about its anterior-posterior axis, the migra- tory pattern of neural crest cells through the sclero- tome is reversed such that they now traverse the posterior (original anterior) halves of the rotated sclerotomes (Bronner-Fraser and Stern, 199 1 ) ( Fig. 3 ) . This pattern is similar to that observed for the navigation of motor axons (Keynes and Stern, 1984). The results suggest that the segmented pat- tern of movement is due to cues inherent to the somite. Thus, there may be inhibitory cues in the posterior sclerotome, attractive cues in the anterior sclerotome. or both.

Inhibitory Effects of the Notochord

Avian neural crest cells migrating along the trunk ventral pathway are distributed throughout the an- terior half of the sclerotome with the exception of a neural crest cell-free space of approximately 85 pm in width surrounding the notochord. In tissue cul- ture experiments, Newgreen found that neural crest cells avoided the region surrounding noto- chords with which they were co-cultured, suggest- ing that the notochord produces a substance that inhibits neural crest migration (Newgreen, Scheel,

Figure 3 Fluorescence photomicrographs of longitu- dinal sections through operated embryos stained with the HNK-1 antibody. ( a ) An embryo in which the seg- mental plate was rotated 180" about its rostrocaudal axis. By the time of fixation, the segmental plate had differentiated into mature somites with dermomyo- tomes and sclerotomes. Neural crest cells are always ob- served in the original rostra1 half of each sclerotome and are absent from the original caudal halves. The arrow indicates a small somite which formed at one of the junc- tions between graft and host tissue. Abbreviations as per Figure I . (From M. Bronner-Fraser and C. Stern, Dev. Biol. 143:2 13-2 17, 199 1 .)

and Kastner, 1986). To determine if the neural crest cell-free space in vivo results from the noto- chord inhibiting neural crest cell migration, we im- planted a length of quail notochord lateral to the neural tube along the neural crest ventral migra- tory pathway of 2-day-old chicken embryos (Fig. 4). The subsequent distribution of neural crest cells was analyzed in embryos fixed 2 days after grafting. When the donor notochord was isolated using collagenase, neural crest cells avoided the ec- topic notochord and were absent from the area im- mediately surrounding the implant (mean distance of 43 pm) (Figs. 4 and 5 ) . The neural crest cell-free space was significantly less when notochords were isolated using trypsin or chondroitinase digestion and was eliminated by fixation of notochords with paraformaldehyde or methanol prior to implanta- tion. These results suggest that the notochord pro-

Page 6: Environmental influences on neural crest cell migration

238 Bronner-Fraser

UNOPERATED OR SHAM EMBRYOS

DISTRIBUTION NEURAL CREST CELLS

EMBRYOS IMPLANTED WITH AN EXTRA NOTOCHORD

- D I STR I BUT I ON NEURAL CREST CELLS

Figure 4 Schematic diagram illustrating the pattern of neural crest cell migration in normal embryos (top) and the effects on neural crest cell migration of implanting an extra notochord adjacent to the neural tube. Abbreviation as per Figure 1.

duces a trypsin and chondroitinase-labile sub- stance that can inhibit neural crest cell migration (Pettway, Guillory, and Bronner-Fraser, 1990). A likely candidate for the inhibitory molecule is a chondroitin sulfate proteoglycan which bears the HNK- 1 epitope (Henning and Schwartz, 199 1 ). The perinotochordal space also appears to be inhib- itory for motor axons (Oakley and Tosney, 199 1 ) .

Dorsoventral Patterning of Neural Crest Derivatives by the Neural Tube

Neural crest cells have a characteristic pattern of migration in the dorsoventral plane. Weston

( 1963) demonstrated that when the neural tube is rotated dorsoventrally by 180" to cause neural crest cells to emerge ventrally, neural crest cells mi- grate in two streams: one dorsally, in reverse direc- tion to that taken normally and the other ventrally towards the aorta. Weston concluded that the neural crest cells are probably not directed to their targets by chemoattractants but rather that they ex- ploit all the spaces available to them. Because these experiments were conducted before it was known that trunk neural crest cells migrating through the sclerotome are restricted to the anterior half, this experimental question was reexamined using mod- ern cell markers.

Page 7: Environmental influences on neural crest cell migration

Netirul Crest Cell Migration 239

Figure 5 Fluorescent photomicrographs of transverse sections through chick embryos im- planted with quail notochords that were isolated by collagenase digestion. The sections were stained with the HNK- 1 antibody, which recognizes a surface epitope on neural crest cells and the perinotochordal matrix. ( a ) An embryo fixed at stage 16. HNK-I immunoreactive neural crest cells approach but do not contact the implanted notochord (arrow), which is located in a ventrolateral position. (b, c ) Low and high magnifications of the same section through an embryo fixed at stage 18 in which the implanted notochord lies lateral to the neural tube. Neural crest cells (curved arrow) appear to be in a ball-like aggregate dorsal to the ectopic notochord (eNo). Abbreviations as per Figure 1. (From Z. Pettway et al., Dev. Bid. 142:335- 345, 1990.)

Using transplantation approaches, we explored the factors that control the polarity, position, and differentiation of the neural crest-derived sympa- thetic and dorsal root ganglion cells. The neural tube, with or without the notochord, was rotated by 180” dorsoventrally to cause the neural crest cells to emerge ventrally. In some embryos, the no- tochord was ablated and in others a second noto- chord was implanted. Neural crest cells emerging from an inverted neural tube migrate in a ventral- to-dorsal direction through the sclerotome, where they become segmented by being restricted to the anterior half of each sclerotome. The dorsal root ganglia always form adjacent to the neural tube, and their dorsoventral orientation often follows the orientation of the grafted neural tube. Simi- larly, the ventral roots emanated from the dorsal portion of the neural tube (originally “ventral” prior to rotation) (Fig. 6) ; however, differentia- tion of sympathetic neurons only occurs near the aorta/ mesonephros and requires the proximity of

either the ventral neural tube or the notochord. The results suggest that the dorso-ventral polarity of the neural tube, dorsal root ganglia, and ventral roots appear to follow the orientation ofthe neural tube. Thus, the neural tube and notochord exert important effects on neural crest cells, influencing the direction of their migration as well as being required for differentiation (Stern, Artinger, and Bronner-Fraser, 199 1 ) .

DISTRIBUTION OF EXTRACELLULAR MATRIX AND ADHESION MOLECULES ALONG NEURAL CREST PATHWAYS

Numerous glycoproteins, proteoglycans, and gly- cosaminoglycans have been described along neural crest migratory pathways. The most prevalent gly- coproteins appear to be fibronectin (Newgreen and Thiery, 1980), laminin (Krotoski et al., 1986), tenascin /cytotactin (Tan, Crossin, Hoffman, and

Page 8: Environmental influences on neural crest cell migration

240 Bronner-Fruser

Figure 6 Fluorescence photomicrographs of transverse sections through an embryo in which the neural tube was rotated 180" in the dorsoventral plane in the absence of a notochord. The embryo was fixed at stage 25, which is well after gangliogenesis normally occurs. The neural tube forms normally but with inverted dorsoventrally polarity. In most sections, the dorsal root ganglia (D) form normally relative to the inverted neural tube. In a few cases, supernumerary dorsal root ganglia form, as illustrated in (c ) . The ventral roots ( V R ) project from the dorsal portion of the neural tube and appear to find their targets in the limb, as seen in (a) and (b). HNK- 1 immunoreactive nerve roots sometimes appeared to cross the midline, as shown in (d) . This is probably due to the absence of the notochord. G = gut; other abbreviations as per Figure 1. Scale bar = 100 pm. (From C. D. Stern et al., Developmerzt 113:207-216, 1991 .)

Edelman, 1987 ) , and various collagens (Duband and Thiery, 1987; Perris et al., 199 1 b). Fibronec- tin, in particular, has been suggested to play a ma- jor role in the adhesion and motility of neural crest cells (Thiery, Duband, and Delouvee, 1982). In tissue culture, neural crest cells migrate avidly on both fibronectin (Rovasio, Delouvee, Timpl, Ya- mada, and Thiery, 1983) and laminin (Newgreen, 1984) substrates; however, the role of fibronectin and laminin in vivo is not yet clear. Of the proteo- glycans present within the embryo, heparan sulfate proteoglycans (Pems et al., 1991b) appear on neural crest cell pathways, whereas chondroitin sulfate proteoglycans are generally present in re- gions from which neural crest cells are absent (Tan

etal., 1987; Pemsetal., 1991b), suchastheperino- tochordal space. Chondroitin sulfate proteoglycans tend to inhibit neural crest cell migration in vitro, consistent with the idea that they may restrict or inhibit migration in the embryo.

The metameric distribution of neural crest cells within the somite may be caused by molecular dif- ferences between the anterior and posterior halves of the somite. A monoclonal antibody discovered by Tanaka and colleagues (Tanaka, Agata, and Obata, 1989) recognizes a molecule in the anterior half of the somite prior to neural crest cell entry into this tissue and whose pattern is unaltered by neural crest cell ablation. Tenascin /cytotactin is also in the anterior half of the somite, although its

Page 9: Environmental influences on neural crest cell migration

N t w u l Crest Cell Migration 241

distribution there appears to require the presence of neural crest cells (Stern et al., 1989; but see Tan et al., 1987, 1991). Other substances have been identified in the posterior half of each somite dur- ing neural crest cell migration, such as molecules bearing a moiety that binds peanut lectin (Stern and Keynes, 1987), a chondroitin sulfate proteo- glycan that binds cytotactin (Tan et a]., 1987) and T-cadherin (Ranscht and Bronner-Fraser, 199 1 ). A functional role for a peanut lectin-binding glyco- protein fraction derived from posterior sclerotome is suggested by experiments in which liposomes containing these molecules inhibit growth cones in vitro (Davies, Cook, Stern, and Keynes, 1990). In addition, collagen types I and 111 and a keratan sul- fate proteoglycan become restricted to the poste- rior half of the sclerotome during advanced stages of neural crest cell migration (Penis, Krotoski, and Bronner-Fraser, 199 1 a). Because the reorganiza- tion of these molecules occurs relatively late, it is likely that they are a consequence, rather than a cause of the neural crest migratory pattern. Thus, the ECM components along neural crest pathways go through dynamic rearrangements at the time of neural crest cell migration.

Because of its presence within the posterior sclerotome, we have examined in detail the distri- bution of T-cadherin (truncated-cadherin), a novel member of the cadherin family of cell adhe- sion molecules, during trunk neural crest cell mi- gration (Ranscht and Bronner-Fraser, 199 l ). T- cadherin was selectively expressed in the posterior half of each sclerotome at all times examined (Fig. 7). T-cadherin immunostaining appeared graded along the anterior-posterior axis, with increasing levels of reactivity in the posterior halves of progres- sively more mature somites. The earliest T-cad- herin expression was detected in a small popula- tion of cells in the posterior portion of the somite three segments anterior to the last-formed somite, concomitant with the initial invasion of neural crest cells into the anterior portion of the same so- mite. When neural crest cells were ablated surgi- cally prior to their emigration from the neural tube, the pattern of T-cadherin immunoreactivity was unchanged compared to unoperated embryos, suggesting that neural crest cells are not required for the metameric distribution of T-cadherin. This distribution is consistent with the possibility that T-cadherin plays a role in maintaining somite po- larity and in influencing the pattern of neural crest cell migration (Ranscht and Bronner-Fraser, 199 1 ) ; however, its functional significance re- mains to be tested.

Figure 7 Fluorescence photomicrograph of a sagittal section through the rostral somites of a stage-1 7 embryo. A double exposure showing that that neural crest cells (red) appear in the rostral half of each somite, whereas T-cadherin (green) appears in the caudal half. Abbrevia- tions as per Figure 1. (From B. Ranscht and M. Bronner- Fraser, Development 11 1: 15-22, 199 1 .)

In contrast to molecules that are selectively dis- tributed within either the anterior or posterior half of the sclerotome, other matrix components in- cluding fibronectin, laminin, and collagen IV re- main uniformly distributed throughout the sclero- tome. Hence, differential distribution of permis- sive and nonpermissive ECM molecules, together with changes in the cells’ ability to interact with the matrix, may determine the migratory pattern of trunk neural crest cells. Although the distribution of extracellular matrix molecules has not been stud- ied as extensively in cranial regions as in the trunk, such molecules as fibronectin, laminin, tenascin, and heparan sulfate proteoglycans are abundant along cranial neural crest pathways (Duband and Thiery, 1987; Krotoski, Domingo, and Bronner- Fraser, 1986; Bronner-Fraser, 1988 ). Perturbation experiments (see below) suggest that these mole- cules may play a functional role in cranial neural crest migration.

Page 10: Environmental influences on neural crest cell migration

242 Bronner-Fraser

ANALYSIS OF CELL-MATRIX INTERACTIONS IN VITRO

Neural crest cells can be grown in vitro by explant- ing neural tubes prior to neural crest migration. They emigrate from the dorsal side of the ex- planted neural tube and form a monolayer on the substrate (Cohen, 1977). Neural crest cells cul- tured in this way migrate avidly on a variety of substrates, including fibronectin (Rovasio et al., 1983; Perris et al., 1989), laminin (Newgreen, 1984; Lallier and Bronner-Fraser, 199 1 ), collagen (Penis et al., 1991a), and other molecules. On fi- bronectin substrates, neural crest cell migration and attachment is progressively enhanced with in- creasing fibronectin concentration. In the case of laminin, however, higher concentrations of sub- strate can actually inhibit cell migration (Perris et al., 1989).

Antibodies that perturb cell-substrate adhesion have been useful for defining the interactions im- portant for cell movement. The CSAT monoclonal antibody recognizes the PI integrin subunit on avian cells (Honvitz, Duggan, Greggs, Decker, and Buck, 1985); in vitro, this antibody inhibits chick neurite outgrowth on laminin, fibronectin, and type-IV collagen (Bozyczko and Honvitz, 1986; Hall, Neugebauer, and Reichardt, 1987; Le- tourneau et al., 1988; Neugebauer, Tomaselli, Li- lien, and Reichardt, 1988). The CSAT antibody against PI integrin can be added to neural crest cells in vitro to test the possible function of the recog- nized epitopes under defined conditions; it disrupts neural crest cell adhesion to fibronectin and laminin substrates (Rovasio et al., 1983; Bron- ner-Fraser, 1985; Lallier and Bronner-Fraser, 199 1 ). The effects are rapid, causing many cells to detach from the dish or aggregate with other cells within 15 min, and are readily reversible upon re- moval of the antibody. These results suggest that binding and dissociation of integrin receptors to matrix molecules occur rapidly. Because not all neural crest cells are affected equally by the anti- body, CSAT may recognize receptors differentially expressed on subsets of neural crest cells or only expressed at certain times. HNK-1 is another cell surface epitope that is present on migrating neural crest cells, neural crest-derived neurons, and some other cells (Tucker et al., 1984). Addition of HNK- 1 antibody to neural crest cells in tissue cul- ture causes detachment and aggregation of the cells grown on laminin substrates ( Bronner-Fraser, 1988).

In order to assess the interactions of neural crest cells with laminin substrates in a quantitative man- ner, we have utilized the cell attachment assay of

McClay ( McClay, Wessel, and Marchase, 198 1 ), which yields highly reproducible results for small numbers of cells. The results show that an increas- ing percentage of neural crest cells adhere to lam- inin with increasing substrate concentrations. This adhesion was inhibited by CSAT antibodies against the chick 0, subunit of integrin, suggesting that &-integrins mediate neural crest cell interac- tions with laminin (Fig. 8) . The HNK- 1 antibody, which recognizes a carbohydrate epitope, inhibits neural crest cell attachment to laminin at low-sub- strate-coating concentrations (> 1 pg/ml; Low- LM) (Fig. 8) , but not at high-coating concentra- tion of laminin ( 10 pg/ml; High-LM) . Surpris- ingly, attachment to Low-LM occurs in the absence of divalent cations. In contrast, attach- ment to High-L,M requires Ca2+ or Mn2’. These results suggest that neural crest cells have at least two integrins that recognize laminin: one that re- quires divalent cations for binding; the other that can function without divalent cations. These ap- pear to recognize different conformations of lam- inin.

We have adapted biochemical techniques uti- lized by von Boxberg, Wutz, and Schwartz, ( 1990) to biotinylate the cell surface of neural crest cells, making it possible to analyze some of their bio- chemical properties for the first time. Using this method, we have performed preliminary biochemi- cal characterization of the HNK-1 epitope on neural crest cells. The HNK- 1 antibody recognizes a 165 kD protein which is also found in immuno- precipitates using antibodies against the /3, subunit of integrin, which precipitates both the 0, and asso- ciated a bands. Our results suggest that the HNK- 1 epitope on neural crest cells is present on or asso- ciated with a novel or differentially glycosylated alpha integrin subunit, which interacts with la- minin in the apparent absence of divalent cations (Lallier and Bronner-Fraser, 199 1 ) . In order to de- termine the identity of this novel 01 subunit, we have obtained various antibodies specific to avian 01 integrin subunits. Interestingly, an antibody against the chick a, subunit of integrin (Syfrig, Mann, and Paulson, 1991) recognizes a 165 kD protein. In preliminary experiments, this antibody inhibits attachment of neural crest cells to laminin in the absence of divalent cations, similar to the effects of HNK-1 antibody.

ANALYSIS OF CELL-MATRIX INTERACTIONS IN VIVO

We have utilized antibody perturbation experi- ments to “knock-out’’ selected cell-matrix interac-

Page 11: Environmental influences on neural crest cell migration

Neural Crest Cell Migration 243

90 --

80

70

60

50

40

30

--

--

--

--

--

--

No Antibody LM

HNK- 1

04 0.1 1 .o 10.0

Substrate Coating Concentration (pg/ml)

Figure 8 Neural crest cell attachment to various concentrations of laminin, ranging from 0.1 to 10 pg/ml, in the presence ofthe CSAT and HNK- 1 antibodies. CSAT and HNK-1 antibod- ies (50 pg/ ml) were added to test their ability to perturb cell adhesion over a range of substrate/ coating concentrations. Points represent the mean of at least six experiments and the error bars represent the S.E.M.

tions along neural crest migratory pathways. These experiments have been performed primarily along cranial neural crest pathways because numerous extracellular matrix molecules are present along these pathways at times that correlate with initial and active migration of this population. Antibod- ies have been introduced along cranial neural crest pathways by microinjection lateral to the mesence- phalic neural tube. Embryos ranging from the neural fold stage to the 9-somite stage are used for injection (Fig. 9). Embryos with greater than 10 somites at the time of injection had no detectable abnormalities, suggesting that they were sensitive to the injected antibodies for only a limited time during their development. After injection, anti- body molecules diffused freely on the injected side of the embryo, but were barely detectable on the uninjected side, as if they did not readily cross the midline.

By microinjecting antibodies that bind and functionally inactivate the P, subunit of the inte- grin, we sought to test the role of the integrin recep- tor in cranial neural crest cell migration in situ. Integrin antibodies caused major defects including reduced numbers of the neural crest cell on the injected side, neural crest cells within the lumen of the neural tube, ectopic neural crest cells external to the neural tube, and neural tube anomalies (Bronner-Fraser, 1985, 1986b). Similar results were obtained with synthetic peptides containing the fibronectin cell binding sequence (Boucaut et al., 1984) or antibodies against fibronectin (Poole and Thiery, 1986). In contrast to the antibodies

that block cell-ECM interactions, several control monoclonal antibodies that bind to integrins but do not block cell-matrix interactions had no detect- able affect on cranial neural crest or neural tube development. These findings support the notion that antibody-induced perturbations in cranial morphogenesis result from a functional block of the integrin receptor and suggest that the receptor

4 - 9 Somite Embryo

\ I I / 1. Inject Ab that blocks

0 1 cell interactions

2. Incubate for 24 -48 hr

3. Section and stain with HNK-1 Ab

Controls: Non-blocking Ab

Figure 9 Schematic diagram illustrating the procedure for injecting antibodies into the cranial mesenchyme ad- jacent to the mesencephalon. Neural crest cells in this region migrate through the mesenchyme underneath the surface ectoderm.

Page 12: Environmental influences on neural crest cell migration

244 Bvonner-Fraser

complex is important in the normal development of the cranial neural crest and neural tube.

The results of experiments using integrin anti- bodies or synthetic decapeptides that competi- tively inhibit cell binding to fibronectin taken to- gether suggest an important function for fibronec- tin in cranial neural crest migration; however, these reagents are not entirely specific. For exam- ple, the cell binding sequence of fibronectin is also present in numerous other extracellular matrix molecules. Likewise, integrin antibodies block neural crest cell adhesion to laminin, fibronectin, and collagens. In addition to using antibodies that interfere with cell binding to fibronectin, we have used an antibody that functionally perturbs cell ad- hesion to laminin as a first attempt to distinguish between the respective roles of these matrix mole- cules.

Laminin is thought to occur in a complex with heparan sulfate proteoglycan (HSPG) in its native state. The inhibitor of neurite outgrowth (INO) antibody recognizes and functionally blocks cell interactions with this laminin-heparan sulfate pro- teoglycan complex (Chiu, Matthew, and Patter- son, 1986). We have injected I N 0 antibody along neural crest pathways in the mesencephalon in order to examine the possible role of laminin in cranial neural crest migration ( Bronner-Fraser and Lallier, 1988). At 1 day after injection, the em- bryos had severe abnormalities in cranial neural crest migration including ectopic neural crest cells external to the neural tube, neural crest cells within the lumen of the neural tube, and neural tube de- formities (Fig. 10). In contrast, embryos injected with antibodies against laminin or heparan sulfate proteoglycan were unaffected. These results indi- cate that functional blockage of a laminin-heparan sulfate proteoglycan perturbs cranial neural crest migration, providing evidence that laminin / HSPG is involved in aspects of neural crest migra- tion in vivo.

Antibodies against several extracellular matrix molecules have been shown to inhibit cranial neural crest migration. In addition to I N 0 and CSAT antibodies described above, antibodies against tenascin (Bronner-Fraser, 1988), the HNK- 1 epitope ( Bronner-Fraser, 1987; which may be present on a subset of integrins), and fibro- nectin (Poole and Thiery, 1986) inhibit cranial neural crest cell migration. In addition to antibod- ies that block cell-matrix interactions, the galacto- syltransferase inhibitor, alpha lactalbumin, in- hibits neural crest cell spreading on laminin, indi- cating a possible role for this enzyme in modulating cell attachment by the alteration of cell

Figure 10 Fluorescence micrograph through the cra- nial region 1 day after injection of I N 0 antibody. Neural crest cells can be recognized by their HNK- 1 immunore- activity. In this embryo, ectopic aggregaters of neural crest cells (arrows) were observed adjacent to the neural tube. The neural tube also appeared deformed. Abbrevia- tion as per Figure 1. (From M. Bronner-Fraser and T. Lallier, J. Ce// Bid. 106:1321-1330, 1988).

surface carbohydrates ( Runyan, Maxwell, and Shur, 1986). Thus, cell-matrix interactions may be multivalent or may occur as an interrelated se- quence of interactions, such that interfering with any step disrupts the process. These results suggest that multiple interactions are necessary for normal migration of cranial neural crest cells and high- lights the fact that complex interactions may be important dunng complicated morphogenetic events.

Interestingly, none ofthe function-blockinganti- bodies that affect cranial neural crest cell migration have been shown to be functional in the trunk re- gion. Because the tissue and extracellular matrix environments appear very different in the head and trunk, it is possible that different strategies may be important for the migration of cranial ver- sus trunk neural crest cells. In the cranial region, it has been established that integrins, laminin, fibro- nectin, and tenascin are involved in some aspects of neural crest migration. In the trunk region, neural crest cell guidance may result from either tissue-derived cues such as differences between the anterior and posterior halves of the somites or in- hibitory substances from the notochord. Thus, cell-matrix interactions may play a permissive rather than an instructive role in the migration of trunk neural crest cells.

CONCLUDING REMARKS

The experiments discussed in this chapter illustrate the important role of interactions between cells

Page 13: Environmental influences on neural crest cell migration

Neural Crest Cell Migration 245

and the extracellular environment in determining the routes of neural crest cell migration. Using a variety of cell marking techniques, it has been possi- ble to document the pathways of neural crest cell migration in the chick and other species. Once the pattern of migration was established, we examined the role of tissue interactions in the patterning of neural crest cell movement. In the trunk region, tissues that line neural crest migratory pathways may dictate the patterns of cell migration. For ex- ample, the metameric pattern of neural crest cell migration through the anterior but not the poste- rior sclerotome appears to be dictated by the so- mites themselves. The absence of neural crest cells from the perinotochordal space appears to arise from an active inhibitory process of the notochord on neural crest cells in its vicinity. In addition, the neural tube is responsible for much of the pattern- ing of neural crest cells along the dorsoventral axis.

Neural crest cells from different regions of the embryo may utilize different mechanisms of cell migration. Both cranial and trunk neural crest cells possess numerous receptors for extracellular ma- trix molecules, including several integrins. In vitro, these integrins mediate neural crest cell attach- ment to fibronectin, laminin, and collagens. Fur- thermore, cell-matrix interactions appear to be re- quired for proper migration of cranial neural crest cells in vivo; however, there is no evidence to indi- cate that a particular cell-matrix interactions is es- sential for dictating the pattern of neural crest cells migration in the trunk region in v i v a Thus, al- though extracellular matrix molecules may serve as a permissive migratory substrate throughout the embryo, they may not necessarily provide specific guidance cues. This suggests that the interactions involved in guidance of neural crest cells are com- plex and often multivalent. The neural crest offers a good experimental system for testing the role of various interactions in cell movement because of the diverse nature of the populations and the range of tissues and molecule with which they can in- teract.

Parts of the work described in this review were sup- ported by USPHS grant HD-15527.

REFERENCES

ARTINGER, K. B. and BRONNER-FRASER, M. E. ( 1992). Partial restriction in the developmental potential of late emigrating neural crest cells in the trunk of the avian embryo. Dev. Biol. 149:149-157.

BRONNER-FRASER, M. (1985). Alterations in neural

crest migration by a monoclonal antibody that affects cell adhesion. J. Cell Biol. 101:610-617.

BRONNER-FRASER, M. (1986a). Analysis of the early stages of trunk neural crest migration in avian em- bryos using the monoclonal antibody €INK-I. Dev. Biol. 115:44-55.

BRONNER-FRASER, M. ( 1986b). An antibody to a recep- tor for fibronectin and laminin perturbs cranial neural crest development in vivo. Dev. Biol. 117528-536.

BRONNER-FRASER, M. ( 1987). Perturbation of cranial neural crest migration by the HNK-I antibody. Drv.

BRONNER-FRASER, M. ( 1988). Distribution and func- tion of tenascin during cranial neural crest develop- ment in the chick. J. Neurosci. Rex 21:135-147.

BRONNER-FRASER, M. and LALLIER, T. (1988). A monoclonal antibody against a laminin-heparan sul- fate proteoglycan complex perturbs cranial neural crest migration in vivo. J. Cell Biol. 106: 132 1 - 1330.

BRONNER-FRASER, M. and STERN, C. ( 199 1 ). Effects of mesodermal tissues on avian neural crest cell migra- tion. Dev. Biol. 143:2 13-2 17.

BOUCAUT, J. C., DARRIBERE, T., POOLE, T. J., AOYAMA, H., YAMADA, K. M . , ~ ~ ~ T H I E R Y , J. P. ( 1984). Biolog- ically active synthetic peptides as probes of embryonic development: a competitive peptide inhibitor of fibro- nectin function inhibits gastrulation in amphibian embryos and neural crest migration in avian embryos. J . Cell Bid . 99: 1822- 1830.

BOZYCZKO, D. and HORWITZ, A. F. ( 1986). The partici- pation of a putative cell surface receptor for laminin and fibronectin in peripheral neurite extension. J . Neurosci. 6: 124 1 - 125 1.

CHIU, A. Y., MATTHEW, W. D., and PATTERSON, P. H. ( 1986). A monoclonal antibody that blocks the activ- ity of a neurite regeneration-promoting factor: studies on the binding site and its localization in vivo. J. Cell Bid. 103:1383-1398.

COHEN, A. M. (1977). lndependent expression of the adrenergic phenotype by neural crest in v i m . Proc. Natl. Acad. Sci. USA 74:2899-2903.

DAVIES, J., COOK, M., STERN, C. D., and KEYNES, R. J. ( 1990). Isolation from chick somites ofa glycoprotein that causes collapse of dorsal root ganglion growth cones. Neuron 4: 1 1-20.

DUBAND, J. L. ~ ~ ~ T H I E R Y , J. P. ( 1987). Distribution of laminin and collagens during avian neural crest devel- opment. Development 101:46 1-478.

HALL, D. E., NEUGEBAUER, K. M., and REICHARDT, L. F. ( 1987). Embryonic neural retinal cell response to extracellular matrix proteins: developmental changes and effects of the cell substratum attachment antibody CSAT. J. Cell Biol. 104:623-634.

HENNING, A. K. and SCHWARTZ, N. B. ( 199 I ). Charac- teristics of a large chondroitin sulfate proteoglycan as- sociated with the notochord in 2- to 4-day old chick embryos. J. Cell Biol. 115:73 la.

HORWITZ, A. F., DUGGAN, K., GREGGS, R., DECKER, C . , a n d B u c ~ , C . ( 1985).TheCSATantigenhasprop-

Biol. 123~32 1-33 1.

Page 14: Environmental influences on neural crest cell migration

246 Bronner-Fraser

erties of a receptor for laminin and fibronectin. J. Cell Biol. 101:2 134-2144.

KEYNES, R. J. and STERN, C. D. (1984). Segmentation in the vertebrate nervous system. Nature 310:786- 789.

KROTOSKI, D., DOMINGO, C., and BRONNER-FRASER, M. (1986). Distribution of a putative cell surface re- ceptor for fibronectin and laminin in the avian em- bryo. J. Cell Biol. 103:1061-1072.

KRUSE, J., MAILHAMMER, R., WENELCKE, H., FAISSNER, A., SOMMER, I., GORIDIS, C., and SCHACHNER, M. ( 1984). Neural cell adhesion molecules and myelin- associated glycoprotein share a common carbohydrate moiety recognized by monoclonal antibodies L2 and HNK- I . Nature 311: 153- 155.

LALLIER, T. and BRONNER-FRASER, M. ( 1988). A spa- tial and temporal analysis of dorsal root and sympa- thetic ganglion formation in the avian embryo. Dev. Biol. 127:99-112.

LALLIER, T. and BRONNER-FRASER, M. ( 1991). Avian neural crest cell attachment to laminin: involvement of divalent cation dependent and independent inte- grins. Development 113: 1069- 1084.

LEDOUARIN, N. M. (1982). The Neural Crest. Cam- bridge University Press, Cambridge.

LETOURNEAU, P. C., PECH, I. V., ROGERS, S. L., PALM, S. L., MCCARTHY, J. B., and FURCHT, L. T. (1988). Growth cone migration across extracellular matrix components depends on integrin, but migration across glioma cells does not. J. Neurosci. Res. 21:286- 297.

LORING, J. and ERICKSON, C. ( 1987). Neural crest cell migration pathways in the chick embryo. Dev. Biol. 121:230-236.

MCCLAY, D. R., WESSEL, G. M., and MARCHASE, R. B. ( 198 1 ). Intercellular recognition: Quantitation of ini- tial binding events. Proc. Natn. Acad. Sci. USA 78:4975-4979.

NEUGEBAUER, K. M., TOMASELLI, K. J., LILIEN, J. , and REICHARDT, L. F. (1988). N-Cadherin, NCAM, and integrins promote retinal neurife outgrowth on astro- cytes in vitro. J. Cell Biol. 107: I 177- 1 187.

NEWGREEN, D. F. ( 1984). Spreading of explants of em- bryonic chick mesenchymes and epithelia on fibronec- tin and laminin. Cell Tissue Res. 3236:265-277.

NEWGREEN, D. F., SCHEEL, M., and KASTNER, V. ( 1986). Morphogenesis ofsclerotome and neural crest in avian embryos: in vivo and in vitro studies on the role of notochordal extracellular matrix. Cell Tissue Res. 244:299-3 13.

NEWGREEN, D. F. and THIERY, J. -P. ( 1980). Fibronec- tin in early avian embryos: synthesis and distribution along the migration pathways of neural crest cells. Cell Tissue Res. 211:269-29 1 .

NODEN, D. M. ( 1975). An analysis of the migratory be- havior of avian cephalic neural crest cells. Dev. Biol.

OAKLEY, R. and TOSNEY, K. W. ( 199 1 ). Peanut aggluti- 42: 106- 130.

nin and chondroitin-6-sulfate are molecular markers for tissues that act as barriers to axon advance in avian embryos. Dev. Bid. 14’7: 187-206.

PERRIS, R., KROTOSKI, D., and BRONNER-FRASER, M. ( 199 1 a) . Collagens in avian neural crest cell develop- ment: distribution in vivo and migration-promoting ability in vitro. Development 113207-216.

PERRIS, R., KROTOSKI, D., DOMINGO, C., LALLIER, T., SORRELL, J . M., and BRONNER-FRASER, M. ( 199 1 b). Spatial and temporal changes in the distribution of proteoglycans during avian neural crest development. Development 111:583-599.

PERRIS, R., PAULSSON, M., and BRONNER-FRASER, M. ( 1989). Molecular mechanisms of avian neural crest cell migration on fibronectin and laminin. Dev. Biol.

PETTWAY, Z., GUILLORY, G., and BRONNER-FRASER, M. ( 1990). Absence of neural crest cells from the re- gion surrounding implanted notochords in situ. Dev. Biol. 142:335-345.

POMERANZ, H. D. and GERSHON, M. D. ( 199 I ). Coloni- zation of the avian hindgut by cells derived from the sacral neural crest. Dev. Biol. 137:378-394.

POOLE, T. and THIERY, J. P. ( 1986). Antibodies and a synthetic peptide that block cell-fibronectin adhesion arrest neural crest cell migration in vivo. In: Progress in Developmentizl Biology, Part B. Alan R. Liss Inc., New York, pp. 235-238.

RANSCHT, B. and BRONNER-FRASER, M. ( 199 1 ). T-cad- herin expression alternates with migrating neural crest cells in the trunk of the avian embryo. Development

RICKMANN, M., FAWCETT, J. W., and KEYNES, R. J. (1985). The migration of neural crest cells and the growth of motor axons through the rostra1 half of the chick somite. J . Exp. Morphol. Emhryol. 90:437.

ROVASIO, R., DELOUVEE, A., TIMPL, R., YAMADA, K., and THIERY, J. P. ( 1983). In vitro avian neural crest cell adhesion and migration: requirement for fibronec- tin in the extracellular matrix. J. Cell Biol. 96:462- 473.

RUNYAN, R. B., MAXWELL, G. D., and SHUR, B. D. ( 1986). Evidence for a novel enzymatic mechanism of neural crest cell migration on extracellular glycocon- jugate matrices. J. Cell Biol. 102:43 1-441.

SERBEDZIJA, G., BRONNER-FRASER, M., and FRASER, S. E. ( 1989). Vital dye analysis of the timing and path- ways of avian trunk neural crest cell migration. Devel- opment 106:806-8 16.

SERBEDZIJA, G., BURGAN, S. E., FRASER, S., and BRON- NER-FRASER, M. ( 199 I ). Vital dye analysis of sacral neural crest cell contribution to the enteric nervous system of chick and mouse embryos. Development

SERBEDZIJA, G., FRASER, S. E., and BRONNER-FRASER, M. ( 1990). Pathways of trunk neural crest cell migra- tion in the mouse embryo revealed by vital dye analy- sis. Development 108:605-6 12.

136~222-239.

11 1: 15-22.

111~857-866.

Page 15: Environmental influences on neural crest cell migration

Neural Crest Cell Migration 247

STERN, C. D., ARTINGER, K. B., and BRONNER-FRASER, M. ( 199 1 ) . Tissue interactions affecting the migration and differentiation of neural crest cells in the chick embryo. Development 113:207-2 16.

STERN, C. D. and KEYNES, R. J. (1987). Interactions between somite cells: the formation and maintenance of segment boundaries in the chick embryo. Develop- ment 99:261-272.

STERN, C. D., NORRIS, W. E., BRONNER-FRASER, M., CARLSON, G. J., FAWNER, A., KEYNES, R. J., and SCHACHNER, M. ( 1989). J 1 /Tenascin-related mole- cules are not responsible for the segmented pattern of neural crest cells or motor axons in chick embryo. De- velopment 107:309-320.

SYFRIG, J., MA", K., and PAULSSON, M. ( 1991). An abundant chick gizzard integrin is the avian a$, inte- grin heterodimer and functions as a divalent cation- dependent collagen IV receptor. Exp. Cell Rex

TAN, S. S., CROSSIN, K. L., HOFFMAN, S., and EDEL- MAN, G. M. (1987). Asymmetric expression in so- mites of cytotactin and its proteoglycan ligand is correlated with neural crest cell distribution. Proc. Natl. Acad. Sci. USA 84:7977-798 1 .

TAN, S. S., PRIETO, A. L., NEWGREEN, D. F., CROSSIN, K. L., and EDELMAN, G. M. ( 199 1 ). Cytotactin ex- pression in the somites after dorsal neural tube and neural crest ablation in chicken embryos. Proc. Natl. Acad. Sci. USA 88:6398-6402.

194~165-173.

TANAKA, H., AGATA, A,, and OBATA, K. ( 1989). A new membrane antigen revealed by monoclonal antibod- ies is associated with axonal pathways. Dev. Biol.

TEILLET, M. A., KALCHEIM, C., and LEDOUARIN, N. M. ( 1987). Formation of the dorsal root ganglion in the avian embryo: segmental origin and migratory behav- ior of neural crest progenitor cells. Dev. Biol. 120:329- 347.

THIERY, J. P., DUBAND, J. L., and DELOUVEE, A. (1982). Pathways and mechanism of avian trunk neural crest migration and localization. Dev. Biol.

TUCKER, G. C., AOYAMA, H., LIPINSKI, M., TURSZ, T., and THIERY, J. -P. (1984). Identical reactivity of monoclonal antibodies HNK- 1 and NC- 1 : conserva- tion in vertebrates on cells derived from neural pri- mordium and on some leukocytes. Cell Differ.

VON BOXBERG, Y., WUTZ, R., and SCHWARTZ, U. ( 1990). Use of the biotin-avidin system for the label- ling, isolation and characterization of neural cell sur- face proteins. Eur. J . Biochein. 190:249-256.

WESTON, J. A. (1963). A radioautographic analysis of the migration and localization of trunk neural crest cells in the chick. Dev. Biol. 6:279-3 10.

WESTON, J. A. and BUTLER, S. L. (1966). Temporal factors affecting the localization of neural crest cells in chick embryos. Dev. Biol. 14:246-266.

132~4 19-435.

93~324-343.

14~223-230.