24
Development 99, 449-471 (1987) Printed in Great Britain © The Company of Biologists Limited 1987 Review Article 449 Oncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA Synopsis Section no. (1) Introduction (2) EGF receptor/c-erb-B (3) CSF-1 receptor/c-/nw (4) c-src (5) c-abl (6) Other oncogene kinases (7) The ras family of genes (8) c-fos (9) c-myb page 449 449 452 452 453 454 454 455 457 (10) The myc gene family (11) c-sis (12) Other proto-oncogenes (13) Proto-oncogene expression in terato- carcinoma cells (14) Conclusions and summary (15) References Kev words: oncoeene. 457 459 460 460 461 462 (1) Introduction Realization that the transforming oncogenes (v-onc) of the acutely oncogenic retroviruses are homologous to cellular genes (and were probably derived from them) brought several areas of research together with exciting prospects for advances in virology, carcino- genesis, evolution and development. The proto-onco- genes (c-onc) are likely to be crucially involved in growth regulation and/or differentiation because of their conservation throughout evolution and because of the well-known growth deregulation effects pro- duced by the \-oncs. It was therefore reasoned that the normal counterpart of these genes should be active during embryonic development and that identi- fication of a specific tissue or stage where c-oncs are expressed should help to identify their roles in all cells and provide a source of material to study the mechan- isms of action. The preliminary results suggest growth modulatory roles for most oncogenes, and develop- mental studies have provided clues to c-onc roles that would not have been forthcoming from studies on cell lines. Several c-onc products have now been identified with specific cellular proteins, and these have con- firmed their importance to growth regulation. They are growth factor or hormone receptors (such as c-erb-B, or epidermal growth factor [EGF] receptor; c-fms, or colony stimulating factor-1 [CSF-1] receptor and c-erb-A, or thyroid hormone receptor) or growth factors (such as c-sis, or B chain of the platelet- derived growth factor [PDGF]). Table 1 lists the proto-oncogenes that have been studied in develop- ing or differentiating systems. For general reviews, see Miiller, 1983; Hunter, 1984; Weinberg, 1984; Varmus, 1984; Heldin & Westermark, 1984; Sin- kovics, 1984; Klein & Klein, 1985; Bishop, 1985; and Muller, 1986. (2) EGF receptor/c-erf>-B The best known of the tyrosine kinase family of c-oncs is the EGF receptor, which is a larger homologue of the v-erb-B protein that causes avian erythroblastosis in chickens infected with avian eryth- roblastosis virus (AEV) (Downward etal. 1984). The EGF receptor has been recognized as a cell-surface glycoprotein whose activity is triggered after binding EGF in the cellular environment. After receptor clustering and endocytosis, lysosomal compartments degrade both receptor and ligand. After this event there is very little known about how the cell receives the signal to commence DNA synthesis and to enter mitosis. Many steps of the process may be necessary, but after the discovery that the receptor also has a tyrosine phosphokinase enzyme activity (Ushiro & Cohen, 1985) that leads to the phosphorylation of itself and of other cellular substrates, the clearest clue

Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Development 99, 449-471 (1987)Printed in Great Britain © The Company of Biologists Limited 1987

Review Article 449

Oncogenes in development

EILEEN D. ADAMSON

La Jolla Cancer Research Foundation, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA

Synopsis

Section no.

(1) Introduction(2) EGF receptor/c-erb-B(3) CSF-1 receptor/c-/nw(4) c-src(5) c-abl(6) Other oncogene kinases(7) The ras family of genes(8) c-fos(9) c-myb

page449449452452453454454455457

(10) The myc gene family(11) c-sis(12) Other proto-oncogenes(13) Proto-oncogene expression in terato-

carcinoma cells(14) Conclusions and summary(15) References

Kev words: oncoeene.

457459460

460461462

(1) Introduction

Realization that the transforming oncogenes (v-onc)of the acutely oncogenic retroviruses are homologousto cellular genes (and were probably derived fromthem) brought several areas of research together withexciting prospects for advances in virology, carcino-genesis, evolution and development. The proto-onco-genes (c-onc) are likely to be crucially involved ingrowth regulation and/or differentiation because oftheir conservation throughout evolution and becauseof the well-known growth deregulation effects pro-duced by the \-oncs. It was therefore reasoned thatthe normal counterpart of these genes should beactive during embryonic development and that identi-fication of a specific tissue or stage where c-oncs areexpressed should help to identify their roles in all cellsand provide a source of material to study the mechan-isms of action. The preliminary results suggest growthmodulatory roles for most oncogenes, and develop-mental studies have provided clues to c-onc roles thatwould not have been forthcoming from studies on celllines.

Several c-onc products have now been identifiedwith specific cellular proteins, and these have con-firmed their importance to growth regulation. Theyare growth factor or hormone receptors (such asc-erb-B, or epidermal growth factor [EGF] receptor;c-fms, or colony stimulating factor-1 [CSF-1] receptor

and c-erb-A, or thyroid hormone receptor) or growthfactors (such as c-sis, or B chain of the platelet-derived growth factor [PDGF]). Table 1 lists theproto-oncogenes that have been studied in develop-ing or differentiating systems. For general reviews,see Miiller, 1983; Hunter, 1984; Weinberg, 1984;Varmus, 1984; Heldin & Westermark, 1984; Sin-kovics, 1984; Klein & Klein, 1985; Bishop, 1985; andMuller, 1986.

(2) EGF receptor/c-erf>-B

The best known of the tyrosine kinase familyof c-oncs is the EGF receptor, which is a largerhomologue of the v-erb-B protein that causes avianerythroblastosis in chickens infected with avian eryth-roblastosis virus (AEV) (Downward etal. 1984). TheEGF receptor has been recognized as a cell-surfaceglycoprotein whose activity is triggered after bindingEGF in the cellular environment. After receptorclustering and endocytosis, lysosomal compartmentsdegrade both receptor and ligand. After this eventthere is very little known about how the cell receivesthe signal to commence DNA synthesis and to entermitosis. Many steps of the process may be necessary,but after the discovery that the receptor also has atyrosine phosphokinase enzyme activity (Ushiro &Cohen, 1985) that leads to the phosphorylation ofitself and of other cellular substrates, the clearest clue

Page 2: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

450 E. D. Adamson

Table 1. Some proto-oncogenes and their protein products

c-oncProtein identity,homology or size

I. Tyrosine kinase and related c-onc

c-erb-B EGF receptor170 K

c-neu

c-fms

c-src

c-mos

c-abl

c-fesc-fps

II. GTPasesc-Ki-raj-2]c-H-nu-1 \

185 KHomology to EGF-R

CSF-1 receptor140 K

60K

37K

150K

92 KJ98 KJ

21K

III Nuclear productsc-fos 55 K

c-myc ^

N-myc 1L-mycJ

c-myb

IV. O^ersc-erfovl

c-iis

62K66K

75 K

T3 receptor,homology toglucocorticoidreceptor

PDGF B chain14 K

Cell location anddistribution

Plasma membrane ofmesodermal, ectodermal& endodermal cells

Plasma membrane

Plasma membrane ofmacrophages & extra-embryonic cells

Cytoplasmic face ofmembranes.Adhesion plaques

Cytoplasmic.Embryotestis & ovary

Plasma membrane

Cytoplasm, plasmamembrane

Membrane cytoplasmicface in a widevariety of cells

Nucleus of extra-embryonic tissues,haematopoietic cells& macrophages Allother cells at lowerlevel.

Nuclear matrix ofmost cells In sometumours & embryonictissues.

Nucleus of haemato-poietic cells

Cytoplasmic &nuclear

Secreted protein

Activities

EGF bindingTGFa bindingTyr. kinase

Tyr. kinase

CSF-1 bindingTry. kinase

Tyr. kinasephosphorylatesmany cellularproteins, e.g.,vinculin, vimentin,filamin, p36.

Ser/thr kinase

Tyr. kinasephosphorylatesvincuhn

Tyr. kinase

GTP + GDPbinding.GTPase.

DNA bindingwith an accessoryprotein

DNA binding

DNA binding

Bindsthyroxine

Homo- orheterodimerbinds toPDGF-R

Possible roles

Signal transductionformitogenesis and differ".Stim° of tooth eruption,eye-opening, lung devel.

Receptor for anunknown ligand?

Signal transduction formitogenesis and differ"

Neurone &muscle development9

B-cell differ1?

Macrophage devel.9

Adenylate cyclaseregulation?

GO to Gl transition.Differentiation

Proliferation.Regulates DNAsynthesis.

Differentiation ofhaematopoietic cells9

Metabolic regulator?

MitogenesisWound healingEarly embryonicgrowth factor?

Chromosomallocationhuman/mouse

7/7

17/?

5/?

20/9

8/?

9/7

15/9

10/711/71/7

14/?

8/15

6/?

17/7

22/7

References

1

2.

3.

4

5.

6.

7

8.

9.

10.

11.

12.

13

K, 10"3iMr

References1. Hayman et al. 1983; Downward el al. 1984; Carpenter 1984. 2. Schechter a al. 1984, 1985; Hung el al. 1986. 3. Rettenmier et al. 1985; Coussens et al.

1986. 4. Purchio et al. 1978; Takeya & Hanafusa, 1983; Sejersenflo/. 1986. 5. Van Beveren et al. 1981; Papkoff«oA 1982, 1983; Baldwin, 1985. 6. Witte «al 1978, 1979, 1980; Reddy a al. 1983. 7. Baibacid et at. 1980; Hampe et al. 1982; Caimier & Samarut, 1986; Ferrari et al. 1985. 8. Tsuchida a al. 1982;Dhar et al. 1982; Hall et al. 1983; MOUer et al. 1982; Cooper a al. 1984. 9. Van Beveren et at. 1983; Deschamps et al. 1985. 10 Mellon et al. 1978; Alitalo etal 1983; Kaczmarek et al. 1985: Eisenman a al. 1985; Wan et al. 1985; Jakobovitz et al. 1985; Zimmerman et al. 1986; Nau et al. 1985. U .Gondana / .1985; Sheiness & Gardinier, 1984; Janssen a al. 1986. 12 HoUenberg et al. 1983; Weinberger a al. 1985, 1986. 13. Devare et al. 1983; Doolittle et al. 1983.

appeared to have been identified. This is because theproportion of phosphotyrosine, compared with phos-phoserine and phosphothreonine in a normal cell, isvery small, about 0-03 % of the total. The mechanism

of oncogenesis via tyrosine phosphorylation has beenpursued vigorously, but the outcome so far has notgiven a clear picture of the roles of tyrosine phos-phokinases in growth regulation. For reviews that

Page 3: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 451

cover the enzymic and metabolic aspects of the EGFreceptor, see Thompson & Gill, 1985; Herschman,1985; Kris, Libermann, Avivi & Schlessinger,1985; Carpenter & Zendoqui, 1986; Soderquist &Carpenter, 1986.

(2.1) The EGF-receptor geneNormal tissues and cells appear to have a single genefor EGF receptor but differential splicing leads to theproduction of at least two mRNAs of about 6 and10 kb (Ullrich et al. 1984; Lin et al. 1984; Xu et al.1984). Both of these appear to code for full-lengthprotein of 170 x 103 Afr (170 K) (Simmen et al. 1984).The gene is amplified in human A431 cells and inseveral types of tumours such as gliomas (Libermannet al. 1984), squamous cell carcinoma (Cowley et al.1984) and retinoblastomas where the protein is alsooverexpressed. Presumably the expression of highlevels of receptor provided a growth advantage to thetumour cells but it is not known if this was the originallesion.

(2.2) Distribution of EGF-binding activity indeveloping tissuesEGF receptors are present on a wide range of celltypes including cells of ectodermal, mesodermal andendodermal origin (reviewed by Adamson & Rees,1981). They are also present on embryonic/fetaltissues and extraembryonic tissues during murinedevelopment (reviewed by Adamson, 1983). But inspite of this, their role in development is not yet clear.EGF receptors increase in number during the ges-tation period so that the new-born's liver expressesmore than any other tissue at any stage (Adamson &Meek, 1984). Crude membrane preparations from allfetal tissues (with the exception of the parietal yolksac) contain EGF-binding activity, and the affinity ofthese receptors for 125I-EGF decreases somewhatduring development. The earliest time of detection ison giant trophoblast cells of a 5-day blastocyst grownfor 2-3 days in culture (Adamson & Meek, 1984).Fetal receptors are functional in that they bind EGFand they can be down-regulated by excess EGFinjected into the placenta or amniotic cavity (Adam-son & Warshaw, 1981). When fetal organs areexplanted and cultured with EGF, incorporation of[3H]thymidine into DNA is stimulated (Adamson,Deller & Warshaw, 1981). Autophosphorylating ac-tivity of EGF receptors is first detected in 10-daymouse embryos at the time of onset of organogenesis(Hortsch, Schlessinger, Gootwine & Webb, 1983).The changing characteristics of EGF receptorssuggests that roles may be different at different stagesof development, although changing cell populationscould also explain changing receptor characteristics in

a tissue such as fetal liver which has high levels ofhaemopoietic cells in the early stages.

Specific tissues have been targets of research aimedto find a role for EGF in their development. EGFstimulates skin differentiation in the neonatal mouseand accelerates eye opening and tooth eruption(Cohen, 1962). Palatal epithelium expresses EGFreceptors that may play a role in palate closure (Tyler& Pratt, 1980; Turley, Hollenberg & Pratt, 1985).Lung development and maturation are affected byEGF (Catterton, Escobedo & Sexson, 1979; Gross etal. 1986; Goldin & Opperman, 1980). Growth ofnewborn rat liver, kidney and craniofacial structuresis retarded by EGF (Hoath, 1986). Chick neural crestcells (Erickson & Turley, 1987), rat oocyte duringmaturation (Dekel & Sherizly, 1985), and postnataland fetal development of rat gastric mucosa (Dem-binski & Johnson, 1985; O'Loughlin et al. 1985;Conteas, DeMorrow & Majumdar, 1986) areinfluenced by EGF.

(2.3) Occurrence of EGF receptors in teratocarcinomacellsThe stem cell lines (embryonal carcinoma, EC) ofmurine teratocarcinomas appear to have very few(F9) or no EGF-binding sites (OC15, PC13, P19).When differentiation occurs, either in aggregate cul-tures (Adamson & Hogan, 1984) or in monolayers(Rees, Adamson & Graham, 1979; Mummery et al.1985), receptors appear which respond to EGF bypromoting cellular proliferation. Teratocarcinoma-derived differentiated cell line PSA5E (visceral endo-derm-like) also has EGF receptors, while PYS andF9AC cells (parietal endoderm-like) do not. Thisdistribution is in agreement with the tissue distri-bution determined from embryo studies. Surpris-ingly, OC15 EC cells have considerable EGF-recep-tor-kinase activity (equivalent to about 3-daydifferentiated cells), but it appears to be largelyintracellular or possibly masked (Weller, Meek &Adamson, 1987). Therefore, we have to modify ouroriginal hypothesis that early embryonic ectodermcells would be negative for this protein based onfindings in EC cells. Indeed, a human EC cell-line(PA1) has been shown to express a low level of EGFreceptors on the cell surface (Carlin & Andrews,1985).

(2.4) Activation of the EGF-receptor oncogeneprotein during developmentIf the EGF receptor is to function it must receivesignals by ligand binding to the extracellular bindingdomain. Two known growth factors bind to the EGFreceptor, EGF/urogastrone and transforming growthfactor a (TGFa). See reviews by Adamson (1983,1986) and Roberts & Sporn (1985).

Page 4: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

452 E. D. Adamson

(2.4.1) EGF

This 53 amino-acid polypeptide (6K) is synthesizedand stored in very large amounts in male adult mouse(but not rat or human) submandibular salivary gland.It is detected in saliva, amniotic fluid, milk, urine andmost tissues of several species. Although EGF hasbeen detected in fetal mice (Nex0, Hollenberg,Figueroa & Pratt, 1980) and fetal rats (Matrisian,Pathak & Magun, 1982), it seems unlikely to bederived from the fetus itself, and Popliker et al. (1986)have presented evidence that the EGF found in fetalmice is derived from the mother. The EGF geneoccurs as a large gene encoding a precursor thatcontains several EGF-like peptides as well as EGF,together with a membrane-spanning domain (Scott etal. 1983). Using a cloned probe for prepro EGFmRNA, Rail et al. (1983) have shown high-level genetranscription in the mouse salivary gland and inkidney. Female kidney expresses about 2- to 4-foldhigher levels than male, and this is a reversal of thesalivary gland levels (Popliker et al. 1986; Gubits etal.1986; Salido et al. 1986). However, only the salivarygland precursor appears to be processed to the active6K form. The kidney 130 K precursor form is notknown to be an active mitogen. The importance ofthese reports is the realization that EGF is notsynthesized until at least 2 weeks postpartum in thekidneys and even later (after weaning) in the malesalivary gland. Then why does the fetus express EGFreceptors?

(2.4.2) TGFa

Rat TGFar (5K) has been purified, sequenced, syn-thesized, molecularly cloned and shown to have35-40 % homology with EGF (Marquardt, Hunkapil-lar, Hood & Todaro, 1983; Tarn et al. 1984; Lee,Rose, Webb & Todaro, 19856). TGFa binds to EGFreceptors and produces exactly the same effects,including mitogenesis and skin maturation. It can alsosynergize with TGF/3 to stimulate anchorage-inde-pendent growth of normal fibroblasts. TGFa alsooccurs as a larger precursor form (Derynck et al. 1984)as a transmembrane protein. TGFa is found innormal rat tissues (Lee et al. 19856; Stromberg,Pigott, Ranchalis & Twardzik, 1982; Proper, Bjorn-son & Moses, 1982; Twardzik, 1985; Massagu6, 1985.In contrast to EGF, TGFa'has been detected as earlyas day 7 in mouse embryos using a specific radioim-munoassay procedure. The levels fall and then rise toa second peak in day-13 fetuses (Twardzik, 1985), inparallel with the levels of mRNA (Lee, Rochford,Todaro & Villareal, 1985a). Later in development,the placenta could be a major source of TGFQ-(Stromberg etal. 1982). These studies imply that earlyembryonic development could be affected by TGFa-binding to EGF receptors, but the details of which

embryonic tissues synthesize the growth factor andwhere TGFa'finds its target are still to be discovered.

(3) CSF-1 receptor/c-frns

The x-fms product is the transforming protein ofthe McDonough strain of the feline sarcoma virus(SM-FeSV). Its proto-oncogene homologue is amembrane-inserted phosphorylated glycoprotein ofapproximately 165 K in mouse and 170 K in cat. Theidentity of the c-fms gene was first suggested by Sherret al. (1985) who showed that it is very similar if notidentical to the monocyte/macrophage cell surfacereceptor for CSF-1. The c-fms gene product is atyrosine kinase, and the gene is therefore a memberof the src family of related oncogenes (Rettenmier,Chen, Roussel & Sherr, 1985; Coussens et al. 1986).

The tissue expression of c-fms appears to be limitedto the monocyte/macrophage lineage (spleen, bonemarrow and fetal liver) and to developing extraem-bryonic tissues in mouse and human where it isexpressed in a stage- and tissue-specific manner(Muller et al. 1983a). Highest levels of accumulatedtranscripts are present in 17th-19th day placenta withlower amounts in amnion, visceral yolk sac andchorion. The question arises whether this expressionin extraembryonic tissues is due to high levels ofmacrophages found there. This will only be answeredwhen in situ localizing methods are performed. How-ever, evidence from a human teratocarcinoma cellline indicates that c-fms may be specific to placentalcells. HT-H cells differentiate spontaneously inmonolayer cultures to trophoblast-like cells that se-crete hCG a sub-units and that express c-fms mRNA(Izhar et al. 1986). This phenotype is cell-type specificsince undifferentiated stem cells and other types ofcells that differentiate from HT-H cells in aggregatesdo not express c-fms and do not secrete hCG. Be Wohuman choriocarcinoma cells also express c-fms,further supporting localization in placental cells(Muller et al. 1983a,6).

(4) c-src

The src proteins were the first tyrosine kinases to bediscovered and form the archetype for this group ofoncogene products, \-src encodes the transformingprotein of the avian Rous sarcoma virus and c-srcencodes a similar 60K protein. The demonstrationthat protein kinases are involved in oncogene-mediated transformation stems from the finding thatpp60"lc is tightly associated with a protein kinaseactivity (Collett & Erikson, 1978; see review by

Page 5: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 453

Wyke, 1983). The Rous SV product also phosphory-lates phosphatidylinositol and diacylglycerol (Sugi-moto, Whitman, Cantley & Erikson, 1984), andso its function is linked with other oncogeneproducts which also affect phospholipid metabolism.A pp60VJnc related tyiosine kinase has been purifiedfrom bovine brain cerebral cortex (Neer & Lok, 1985)as a 61K protein that can be phosphorylated on serineas well as tyrosine. It exhibits an autophosphorylatingactivity and also phosphorylates precipitating anti-body (Resh & Erikson, 1985). Its location is on thecytoplasmic face of the cell as well as perinuclear. Theamino terminus of pp6ffrc is myristylated, and this isneeded both for localization to the inner face of theplasma membrane and for cellular transformation(Kamps, Buss & Sefton, 1985).

When the pp60v~jrc gene is transfected into NIH3T3 cells, gap junction communication between ad-jacent cells is inhibited and the activity of proteinkinase C is enhanced (Chang et al. 1985). Theseactivities suggest that if c-src has a similar function, itcould be important to development. It was foundseveral years ago that chick embryonic neural tissuesexpress high levels of c-src (Cotton & Brugge, 1983).Immunoassays show that most tissues have eight- toten-fold lower levels of c-src than brain, retina andspinal ganglia. Immunocytochemical studies showhighest levels in neural tube, brain and heart of stage-32 chicks, with lower levels in eye, limb bud and liver.A similar distribution is found in human fetuses withhighest levels in cerebral cortex, spinal cord and heart(Levy, Sorge, Meymandi & Maness, 1984; Gessler &Barnekow, 1984). Thus the expression of c-src corre-lates strongly with the differentiation of electrogenictissues when proliferation has ceased and expressionpersists in terminally differentiated neurones.

Recent immunocytochemical staining results ofManess, Sorge & Fults (1986) show that developingneural tissues in chick exhibit pp6O*rc expression attwo different stages. On or before stage 4, transientlocalization is seen in the neural ectoderm. Thisdeclines by stage 12 (45-49 h) and later rises again interminally differentiated neurones at about stage 21(day 3-5) in the neural retina (Sorge, Levy & Maness,1984) and stage 17 (day 2-5) in the cerebellum (Fults,Towle, Lauder & Maness, 1985). Therefore it ap-pears that c-src may play a role at proliferative stagesalso. An analogous pattern of c-src expression inDrosophila has been shown by in situ hybridization(Simon, Drees, Kornberg & Bishop, 1985). c-srcmRNA is abundant in early embryos during gastru-lation, low in larvae, and high in neural tissue andsmooth muscle and pupae at later stages.

In vitro culture systems have been used to elucidatethe location, effects and roles of c-src. Immunostain-ing shows that c-src is distributed all over cell bodies,

processes and growth cones of chick dorsal rootganglion cultures (Maness, 1986). Primary cultures ofneurones or astrocytes from rat brain contain 15- to20-fold more c-src protein than fibroblasts, and thekinase specific-activity of c-src in neurones is 6- to 12-fold higher than in astrocytes (Brugge et al. 1985).Intriguingly, neuronal but not astrocytic c-src differsin size by a post-translational modification in theamino-terminal half. The murine teratocarcinomacell line, PCC7, can be induced to differentiate intoneuronal structures and a parallel elevation (3- to 5-fold) in c-src mRNA can be detected (Sejersen,Bjorklund, Sumegi & Ringertz, 1986). An 8- to 20-fold increase in src protein levels is observed after 5days of induction of P1951801A1 murine EC cellswith retinoic acid, corresponding to the appearanceof neuritic processes and other neuronal markers.The induced src protein is the slower mobility formassociated with neurones, a finding that suggests theusefulness of teratocarcinoma cell model systems(Lynch, Brugge & Levine, 1986).

Studies with RSV infections or \-src introducedinto cultured cells have also given results that suggestboth proliferation and differentiation can be inducedby the gene. For instance, marrow cultures areinduced to greater selfrenewal of haematopoieticprogenitor cells (Boettiger, Anderson & Dexter,1984), while retrovirus carrying \-src introduced intoPC12 rat phaeochromocytoma cells induces somefeatures characteristic of differentiation (neurite ex-tension) (Alema, Casalbore, Agostini & Tato, 1985).v-src-containing viruses suppress differentiation inchondroblasts as measured by the synthesis of chon-drocyte-specific products (Alema, Tato & Boettiger,1985). However, \-src has different properties to c-srcand may also be regulated differently.

(5) c-abl

The Abelson leukemia virus (Ab-MLV) induces Band T cell lymphomas in mice by means of theexpression of v-abl, which encodes a tyrosine kinaseof 160 K. Cooperation with EGF receptors appears tobe necessary for v-abl to transform murine fibroblastsinto tumorigenic cells (Gebhardt, Bell & Foulkes,1986). An homologous c-abl of 150 K is found innormal mouse cells (Goff, Gilboa, Witte & Balti-more, 1980). c-abl is actively transcribed duringembryo and fetal development with high levels oftranscripts in extraembryonic tissues as well as in thefetus proper (Miiller et al. 1982). Its expression ishighest on day 10 when organogenesis is progressingrapidly. Thereafter, mRNA levels fall during ges-tation but are detectable throughout. Low levels havebeen detected in several teratocarcinoma cell lines(Sejersen, Sumegi & Ringertz, 1985).

Page 6: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

454 E. D. Adamson

(6) Other oncogene kinases

Very little is known about the expression of the othertyrosine kinases in development. The proto-onco-gene c-ros has features in common with the EGFreceptor family and displays tissue-specific and de-velopmentally regulated expression (Neckameyer,Shibuya, Hsu & Wang, 1986). The oncogene \-kit,which is the transforming gene of HZ4 feline retrovi-rus, has homologous regions to PDGF receptor andc-fms, but is presumably a truncated version with notransmembrane domain (Besmer et al. 1986).

Distantly related to the tyrosine kinase encodingoncogenes is v-raf, which has ser/thr kinase activity.The c-raf-1 gene has been cloned and utilized toevaluate expression in mouse embryos. mRNA is notdetected, while a related but distinct gene, A-raf, istranscribed in 14-day embryos with less in 18-dayembryos and placenta. Moderate levels were found incertain adult tissues (Huleihel et al. 1986).

The cellular homologue of the transforming geneproduct, p2>lc'mos, of the Moloney murine sarcomavirus (Papkoff, Nigg & Hunter, 1983) was thought tobe unexpressed in mouse tissues. By sensitive SInuclease and Northern assays, expression has beendetected in whole embryos at moderate levels, inplacenta, kidney and brain at very low levels, and athigh levels in adult testes and ovary. Transcript sizesdiffer in different tissues and it was suggested thattissue-specific regulation of the size of mos transcriptscould be transactivated, for example, by hormones,and could give rise to functionally different proteinproducts (Propst & Vande Woude, 1985).

(7) The ras family of genes

The ras genes were first identified as the viral onco-genes of the Harvey and Kirsten rat sarcoma viruses(Ellis, DeFeo, Furth & Scolnick, 1982). Cellular rasgenes have been identified in most species, and theyconstitute a family of three human genes whichencode a remarkably well-conserved protein, desig-nated p21 (Defeo et al. 1981). The p21 protein islocated on the cytoplasmic face of the plasma mem-brane in both normal and transformed cells. In vitro,p21 binds GTP (Finkel, Der & Cooper, 1984), but thenormal gene product hydrolyses GTP at a rate 8- to10-fold higher than that of the transforming protein(Sweet et al. 1984). ras proteins are structurally andfunctionally analogous to the G proteins which areinvolved in adenylate cyclase regulation (Gilman,1984; Hurley et al. 1984). The potential relationshipbetween adenylate cyclase and p21 may be part of thecontrol of cell division in Xenopus laevis oocytes.Progesterone-induced meiotic activation of germinalvesicle breakdown (GVBD) is mediated, at least in

part, by inhibition of the oocyte adenylate cyclase(Finidori-Lepicard et al. 1981), but it appears not toinvolve the inhibitory guanine-nucleotide bindingsubunit G,. Sadler, Schechter, Tabin & Moller (1986)showed that monoclonal antibodies to p21-ras in-hibited adenylate cyclase activity and gave acceler-ated maturation of Xenopus laevis oocytes in a dose-dependent manner. It is possible either that p21protein interacts with the pathway of normal celldivision regulated by progesterone or that antibodiescross react with the oocyte G proteins. Ras proteinsappear to interact with phospholipase C in a Gprotein-like manner (Fleischman, Chahwala & Cant-ley, 1986), and c-Ki-ras-2a has some homology withlipocortin and related proteins (Kretsinger & Creutz,1986).

The ras gene products are clearly involved in animportant aspect of cell proliferation since evennormal p21c""" when expressed at elevated levels canresult in the immortalization and transformation ofmouse cells (but not human cells). c-Ha-ras and c-Ki-ras expression is detected at all stages of mouseembryonic development at almost unvarying levels(Miiller et al. 1982; Muller, 1983; Slamon & Cline,1984). This would suggest a general metabolic role indevelopment, ras proteins are remarkably conservedevolutionarily, with homologous proteins occurringin yeast which have similar GTP-binding and hydro-lytic properties (Temeles et al. 1985) and inDrosophila (Mozer, Marlor, Parkhurst & Corces,1985). Of the three v-Ha-ras-related cellular genes inD. melanogaster, each is transcribed into two sizes ofmRNAs. The larger is expressed at similar abundanceduring the life cycle stages, while the shorter tran-script is more abundant in embryonic stages (Lev,Kimchi, Hessel & Segev, 1985). In situ hybridizationwas used to locate and identify active tissues. Distri-bution is uniform in embryos but is restricted todividing cells in larvae. However, in the adult, bothdividing and nondividing tissues contain high levels ofras transcripts, including ovaries, cortex of brain andganglia, thus suggesting roles in growth and in differ-entiation (Segal & Shilo, 1986). An homologous p23protein in Dictyostelium discoideum is expressed athighest levels in growing organisms, and this declineswith the onset of differentiation (Pawson et al. 1985).Similarly, F9 murine teratocarcinoma cells expresshigh levels of c-Ha-ras mRNA and this is moderatelydiminished during differentiation (Campisi et al.1984). However, c-Ki-ras expression increases after48 h of stimulation of differentiation of mouse eryth-roleukemic cells with DMSO. Other oncogenes arealso induced, including fos, myb, and myc, while tenothers remain unchanged (Todokoro & Ikawa, 1986).

The differentiation inducing properties of c-Ha-raswere tested by introducing sarcoma viruses carrying

Page 7: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 455

ras oncogenes into PC12 cells (Noda etal. 1985) or bymicroinjecting purified normal or activated Ha-rasproteins (Bar-Sagi & Feramisco, 1985). There is noeffect of normal c-ras on PC12 differentiation, butactivated Ha-ras products induce differentiation inboth cases. Antibody to p21-ras protein microinjectedinto PC12 cells inhibits nerve-growth-factor-induceddifferentiation (Hagag, Halegoua & Viola, 1986),and this indicates that c-ras does indeed play a role indifferentiation.

The expression of c-Ki-ras appears to be cell cycledependent in a chemically transformed mouse fibro-blast cell line with highest expression in mid to lateGo/Gi (Campisi etal. 1984). In addition, antibody toras microinjected into quiescent NIH 3T3 cells priorto serum stimulation blocks a large population of cellsfrom entering S phase later while control antibodiesdo not. A time course shows that c-ras activity isneeded just before S phase or about 8 h after additionof serum (Mulcahy, Smith & Stacey, 1985). A similartime of c-Ha-ras gene activation is found after partialhepatectomy in rats, and this returns to normal after 3days suggesting that proto-oncogene regulation is anormal regulated process in non-neoplastic growthprocesses (Goyette, Petropoulos, Shank & Fausto,1984).

(8) c-fos

The FBJ and FBR murine osteosarcoma virusesrapidly induce osteosarcomas in mice, and a 55 Kprotein encoded by the transforming gene, v-fos, ofthe FBJ virus (J5&a*-fos from FBR-MSV) has beendescribed (Curran & Teich, 1982; Curran & Verma,1984). The cellular homologue, c-fos, also encodes a55 K protein, which differs from the viral protein inthe carboxy-terminal portion, but which, neverthe-less, can transform normal fibroblasts (Miller, Curran6 Verma, 1984). Both proteins are nuclear phospho-proteins and both are turned over rapidly in the cell,\-fos with a half life of 2 h and c-fos about 20 min. c-fosprotein differs in the degree of post-translationalmodifications that can be detected soon after syn-thesis (Curran, Miller, Zokas & Verma, 1984). Seereviews by Miiller & Verma (1984) and Deschamps etal. (1985) for further details.

(8.1) c-fos expression in developing tissues andproliferating cellsA distinguishing feature of c-fos expression is thatalthough transcripts are present at barely detectablelevels in embryos and fetuses, the extraembryonictissues have very high levels (Miiller etal. 1982). Day-7 murine conceptuses consisting predominantly ofextraembryonic tissues have very high levels of ex-pression and, on further examination, a stage- and

tissue-specific pattern is detectable (Muller, Verma &Adamson, 1983c). In the mouse, amnion >visceralyolk sac > placenta, and a similar pattern is found inhuman tissues (Muller etal. 19836). In the mouse thelevel of c-fos mRNA rises to a plateau on the 16th to17th day of gestation in extraembryonic tissues andalso 14th day (haematopoietic) fetal liver, and laterskin and bone/bone marrow contain high levels.Fetal liver and bone marrow probably express c-foslargely because of the population of macrophages andother haematopoietic lineages that express c-foseither constitutively or at some stage in their differen-tiation/maturation processes (Muller, Muller & Guil-bert, 1984; Muller, Curran, Muller & Guilbert, 1985).Macrophages cannot account for the high expressionin very early extraembryonic tissues. In situ localiz-ation of c-fos protein (Adamson, Meek & Edwards,1984) and mRNA (Deschamps et al. 1985) has clearlylocated fos expression in all the cells of the extraem-bryonic tissues. In addition, extraembryonic tissueshave been shown to synthesize p55c"^OJ protein(Mason, Murphy & Hogan, 1985). Therefore, what isthe role of c-fos gene expression in these tissues?

A brief survey follows of the three types of stimulithat induce c-fos expression, including growth, differ-entiation and stress stimuli. Quiescent, serum-starvedmouse or rat fibroblasts in Go, stimulated to 'com-petence to divide' by PDGF, FGF (or serum), acti-vate the fos gene within a few minutes; mRNA levelspeak at 30 min and then fall to low levels in 60 min.This is caused by an increased rate of transcriptionand is accompanied by increased protein synthesis.The increased levels of mRNA and protein synthesisare both transient (Greenberg & Ziff, 1984; Muller,Bravo, Burckhardt & Curran, 1984; Kruijer, Cooper,Hunter & Verma, 1984; Treisman, 1985; Zullo, Coch-ran, Huang & Stiles, 1985; Renz et al. 1985; Green-berg, Hermanowski & Ziff, 1986). In all cases c-fosmRNA accumulations are followed by an increase inthe level of c-myc mRNA, although it is not known ifthese are linked responses. Epithelial and other cellsrespond to their specific mitogens with a similartransient increase in fos mRNA: EGF-stimulatedA431 cells (Bravo, Burckhardt, Curran & Muller,1985); PC12 cells (Greenberg, Greene & Ziff, 1985);and primary hepatocytes (Kruijer etal. 1986); thymo-cytes stimulated with concanavalin A (Moore, Todd,Hesketh & Metcalf, 1986); thyroid cells with thyro-tropin (Colletta, Cirafici & Vecchio, 1986; Tramon-tano, Chin, Moses & Ingbar, 1986); peripheral lym-phocytes with phytohaemagglutinin and calciumionophore (Reed, Alpers, Nowell & Hoover, 1986).Primary amnion cell cultures continue to express c-fosprotein for 2h after culture in vitro but this leveldeclines to zero in 15 h. Addition of undefined factorsin medium conditioned by placental or embryo

Page 8: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

456 E. D. Adamson

explants stimulates the re-expression of fos protein(Miiller et al. 1986). Apparently, constitutive ex-pression of fos in amnion may be maintained bystimulatory endogenous factors that are produced byfetal and placental tissues. These examples suggestthat c-fos is transiently expressed whenever mitogensbind to a cell receptor, but the following cases showthat the subsequent response of a cell is not necess-arily correlated with cell division.

(8.2) c-fos induction associated with celldifferentiationWhen PC12 cells are stimulated with nerve growthfactor (NGF), they sprout neurites and becomedifferentiated, non-dividing, neurone-like cells. Thec-fos gene is rapidly activated with similar kinetics aswhen proliferation is stimulated with EGF or FGF(Greenberg et al. 1985; Kruijer, Schubert & Verma,1985; Milbrandt, 1986). In contrast, c-fos is notactivated when PC12 cells differentiate to chromaf-fln-like cells when treated with glucocorticoids(Deschamps et al. 1985). In the case of PC12 cells,c-myc is also activated, whereas in other differen-tiating systems c-myc mRNA levels usually fall pre-ceding or accompanying falling levels of prolifer-ation. Other cell types that express elevated c-fosmRNA levels when stimulated to differentiate in-clude HL-60 and U-937 human leukemia cells whentreated with the phorbol ester TPA (Mitchell, Zokas,Schreiber & Verma, 1985; Miiller et al. 1985) andWEHI-3B cells induced with granulocyte-colonystimulating factor (Gonda & Metcalf, 1984). In thelatter case, c-fos mRNA accumulates to high levelsonly after 2-3 days, when differentiation to mono-cytes occurs. In cell lines that differentiate to mono-cyte/macrophages,/as mRNA levels remain modera-tely high and therefore differ from growth factorresponses. The activation of c-fos appears to belineage-specific since HL-60 cells differentiating togranulocytes do not activate fos (Miiller et al. 1985;Mitchell et al. 1985). c-fos expression is constitutivelyhigh in a mast cell precursor line (PB-3c) grown in thepresence of interleukin 3 (IL-3) (Conscience, Verrier& Martin, 1986). When IL-3 is withdrawn, the c-foslevel falls and is rapidly induced when growth factor isadded back. This cell line and macrophages expresshigh levels of fos while actively proliferating asdifferentiated cells. It cannot be concluded thatcertain lineages of differentiation require fos acti-vation, since an HL-60 cell line resistant to TPA canbe induced to differentiate to macrophages in theabsence of detectable c-fos mRNA expression (Mit-chell, Henning-Chubb, Huberman & Verma, 1986).Neither can it be concluded that fos is not requiredsince it is possible that a stable form of fos protein ispresent.

(8.3) c-fos activation by other stimuli such as heatshockFrom the above examples, it can be seen that manydifferent external stimuli seem to activate the tran-sient expression of c-fos mRNA. When quiescentHeLa cells are changed from culture at 37 °C to 40 °Cto 44°C, a temperature-dependent, 5- to 20-foldincrease in c-fos mRNA and protein levels follows inabout lh (Andrews, Harding, Calbet & Adamson,1987). Ionic signals such as calcium ionophore, ben-zodiazepines and elevated K+ (Curran & Morgan,1985; Morgan & Curran, 1986), the cell divisioninhibitor mitomycin C and ultraviolet light (unpub-lished observations of S. Edwards and E. Adamson)all induce c-fos mRNA expression. Partial hepa-tectomy and even a sham operation elevate c-fosmRNA levels in rat liver (Kruijer et al. 1986).Wounding a fibroblast monolayer results in rapidinduction of c-fos (Verrier, Miiller, Bravo & Miiller,1986). /3-adrenergic stimulation of mice in vivo pro-duces hyperplasia in parotid and submandibular sali-vary glands and stimulates transient c-fos expression(Barka, Gubits & Vander Noen, 1986). In summary,many kinds of external stimuli achieve a rapid cellularresponse in the form of c-fos gene induction andstrongly suggests that fos is important in relayingextracellular signals to the nucleus, but the mechan-isms remain unknown.

(8.4) c-fos expression in teratocarcinoma modelsystems of developmentc-fos mRNA is expressed at very low levels inproliferating EC cells (Miiller, 1983; Vilette,Emanoil-Ravier, Tobaly & Peries, 1985; Edwards &Adamson, 1986) and increases modestly during F9aggregate differentiation to embryoid bodies, peak-ing on day 3 after retinoic acid addition (S. Edwardsand E. Adamson, unpublished data; Miiller, 1983).P19 EC aggregates stimulated with DMSO to differ-entiate into a mixture of cell types, including cardiacmuscle express c-fos mRNA in increasing amountspeaking on the 12th day (Edwards & Adamson,1986). These results indicate that c-fos expression canaccompany teratocarcinoma cell differentiation andsupport the findings of Miiller & Wagner (1984) andRuther, Wagner & Miiller (1985) who showed thatthe integration and expression of normal exogenousc-fos genes in F9 cells stimulates differentiation. P19EC cells are less affected, however, and PC13 cellsare not stimulated to differentiate by c-fos expression.Thus c-fos expression alone is not sufficient to triggerdifferentiation. Apparently contrary to the hypoth-esis that c-fos expression may be necessary (but notsufficient) for differentiation to occur, Mason et al.(1985) did not find elevated c-fos mRNA levels duringF9 differentiation in monolayer cultures stimulated

Page 9: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 457

with retinoic acid and cAMP, but did detect atransient slight induction 60min after RA addition.

An effective way to determine the function of agene is to introduce complementary RNA into cellsand determine the effect of the prevention of theexpression of the protein product of that gene. Theintroduction of 'anti-sense' fos DNA into 3T3 cellshas already shown to be effective in reducing growthrates and in reducing the frequency of such clones(Holt, Gopal, Moulton & Nienhuis, 1986). Ourunpublished results indicate that anti-sense fos DNAexpressed in F9 EC cells inhibits their ability todifferentiate in response to RA. This experimentalapproach together with antibody injections, willlikely become prominent in the near future. So far,the data support a hypothesis that fos may be import-ant to development in two ways: one, to act as the'second messenger' for a variety of external stimuliand second, to act in some part of the differentiationprocess, perhaps a step that is connected with growthrestriction or with initiating a new programme of generegulation associated with differentiated function.

(9) c-myb

c-myb is the homologue of the viral transforming genein E26 and other avian viruses that produce myelo-blastosis in birds, c-myb expression is restricted to,and is developmentally regulated, in haematopoietictissue and is therefore thought to play a specific rolein haematopoiesis (Duprey & Boettiger, 1985). c-mybis expressed in a differentiation stage-specific mannerin pre-B cells lines (DeCino, Herbst, Lernhardt &Raschke, 1987). Five percent of the haematopoieticcells of the chick yolk sac contain all the detectablec-myb mRNA of that tissue. These cells were ident-ified as M-CFC or the committed progenitors to themacrophage lineage. As the cells differentiate, thelevel of c-myb falls more than 100-fold. A similar fallaccompanies WEHI-3B cell differentiation to macro-phages (Gonda & Metcalf, 1984). Proto-oncogenec-myb expression is not restricted to macrophagessince both T cells in the murine thymus and cells ofthe erythroid lineage also express c-myb, and this fallstenfold with increasing age (Sheiness & Gardinier,1984). In situ hybridization confirms these lineagesand demonstrates the presence of c-myb mRNA inrapidly proliferating precursors of the myeloid anderythroid lines in human bone marrow cells (Emilia etal. 1986).

(10) The myc gene family

c-myc is the genomic counterpart of the transforminggene of the avian myelocytomatosis virus (MC29)

and, like c-myb and c-fos, its protein product movesrapidly to the nucleus after synthesis and is short-lived. Other homologous genes have been termedL-myc and N-myc corresponding to their activitydetected in lung and neural tumours, respectively.The examples of c-myc expression in response tomitogens of many kinds have been listed in thesection on c-fos and include thrombin, with insulinacting on Chinese hamster fibroblasts (Blanchard etal. 1985). In contrast, there are some exceptionswhere growth-stimulating factors do not induce c-mycmRNA levels: insulin (Kelly, Cochran, Stiles &Leder, 1983); B cell growth factor (Smeland et al.1985), and adenovirus infection (Liu, Baserga &Mercer, 1985). In spite of the large variations seen inc-myc mRNA levels, its rate of transcription increasesonly modestly when cells move from Go to Gi andthen remains at the same lower level in all phases ofthe cell cycle (Thompson, Challoner, Neiman &Groudine, 1985; Rabbitts et al. 1985; Kaczmarek,1986) and differentiation (Dean, Levine & Campisi,1986). Even more than for c-fos, post-transcriptionalregulation is the major means by which the level ofexpression of c-myc is modulated. However, forgrowth stimulation of fibroblast cells by growth fac-tors, an elevated transcription rate can occur depen-ding on the cell type and the growth factor.

(10.1) c-myc expression and cell proliferationc-myc, until recently, has been well correlated tovarious aspects of cell proliferative states (Birnie,Burns, Clark & Warnock, 1984). In transgenic mice,where levels of c-myc are elevated by introduction ofthe c-myc proto-oncogene coupled to the immuno-globulin [i or K enhancer, frequent occurrence oflymphomas occurs within a few months of birth(Adams et al. 1985). Translocation of the c-myc geneto a chromosomal location that endows altered ex-pression has been found in many leukemias in mouseand human. Although induction of the c-myc geneoccurs when erythroleukemic cells are stimulated todifferentiate, expression is transient and, in general,low levels of c-myc are present in differentiatinghaematopoietic cells (Gonda & Metcalf, 1984; Lach-man & Skoultchi, 1984). In view of the association ofc-myc with proliferation, it is not easy to understandwhy both mitotic and meiotic phases of germ cellshave very low levels of c-myc transcripts (Stewart,Bellve & Leder, 1984). For somatic cells likequiescent Swiss 3T3 fibroblasts, DNA synthesis isstimulated by c-myc protein injected into the nuclei(Kaczmarek et al. 1985). Here c-myc acts like acompetence growth factor to initiate the cell cycle sothat cells progress into S phase. It now appears that atleast one way that c-myc may act is by participating inthe process of DNA synthesis, since the addition of

Page 10: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

458 E. D. Adamson

affinity-purified anti-c-myc antibodies to isolated nu-clei from several types of human cells reversiblyinhibits DNA synthesis and DNA polymerase activityof the nuclei (Studzinski, Brelvi, Feldman & Wall,1986).

(10.2) c-myc in developmentNot surprisingly, the c-myc gene is highly activeduring development. Stage-specific expression ofc-myc mRNA was found by Pfeifer-Ohlsson et al.(1984) in human placenta with 30-fold variation inlevel. Four- to five-week placenta was the highest,and this declines to much lower levels by the end ofthe first trimester. The mRNA is located predomi-nantly in the cytotrophoblast cells where [3H]thymi-dine labelling also shows these cells to be rapidlydividing. The nondividing syncytiotrophoblast of theterm placenta has 40-fold lower levels. Interestingly,not every cytotrophoblast cell contains c-myc tran-scripts, and it was suggested that a wave of mitoticactivity moving down the placental villi may correlatewith accumulated levels of c-myc mRNA. A mostrelevant observation is that coexpression of c-sis andc-myc occurs in the early placenta (Goustin et al.1985). Since c-sis codes for a PDGF-like mitogen,secretion of this activity into the medium of culturedtrophoblasts of first trimester placenta may not becoincidental (see section 11).

The human fetus proper also displays stage- andcell-specific expression of c-myc with highest levels inthe rapidly proliferating epithelial tissues. These highlevels are sustained throughout the first trimester incontrast to declining levels in the placenta during thesecond month of gestation. The distribution of c-myclevels implies more than mere association with pro-liferation; in 3- to 4-week embryos, c-myc expressionis low, while in extraembryonic tissue c-myc mRNAlevels are high (Pfeifer-Ohlsson et al. 1985). Further-more, predominantly normal development occurs intransgenic mice that express varying levels of c-myc(Leder et al. 1986).

During murine development, c-myc mRNA hasbeen observed at high levels throughout, but a relatedgene, N-myc, is highly expressed at very early stages(7-5 days of gestation) and continues at this level until11-5 days when it declines thereafter (Jakobovits,Schwab, Bishop & Martin, 1985). Zimmerman et al.(1986) examined the expression of all three myc-reiated genes in murine postnatal development andobserved that c-myc expression is present at all stagesand in all tissues, c-myc levels decline to low levels inolder adults in most tissues except adrenals, thymus,spleen, intestine and heart. Tissue- and stage-specificexpression was demonstrated for L-myc and N-myc.L-myc expression is highest in newborn forebrain,hindbrain and kidney, at lower levels in lung and

intestine, and absent in other tissues. L-myc ex-pression is still present in adult lung but decreases inall other tissue so that a 17-day-old brain no longerexpresses L-myc. N-myc was thought to be neuroecto-derm-specific but in fact is found in a variety ofnewborn tissues. It is highest in newborn brain,kidney and intestine, and declines rapidly with in-creasing age. In addition, a striking differential distri-bution is observed in various B cell lines. N-myc ispresent in pre-B cells but not B cells or plasma cells,while c-myc is expressed in all of these lines, and L-myc is never expressed. Very high c-myc expressionoccurs in late fetal mouse cerebellum that declinesand is succeeded by a second peak in postnatal days 3to 10 (Ruppert, Goldwitz & Wille, 1986). The latter islocalized in the mitotically active external granularlayer and accompanies a change in the ratio of the twomyc mRNAs to adult ratios. In general, the ex-pression of myc family genes correlates with commit-ted but still proliferating cells as well as with cells thatare rapidly differentiating along the neural pathway.

The only non-neural tissue in which a relativelyhigh level of expression of N-myc and c-myc isobserved is fetal and newborn kidney. If the mycfamily genes can all be stimulated by growth factors,possibly either the production and activity of prepro-EGF accounts for myc activation, or more likely, theconstant exposure of the kidney to blood-bornegrowth factors stimulates high levels of expression ofthese oncogenes. N-myc is also expressed in mouseand human teratocarcinoma stem cells and in adultmouse testis (Tainsky, Cooper, Giovanella & VandeWoude, 1985; Jakobovits et al. 1985).

The myc gene is highly conserved through evol-ution and even Drosophila melanogaster genome con-tains sequences that hybridize with the v-myc probe,although there is little amino acid sequence homologyin the products. Nevertheless, hybridizing transcriptsare found in embryos, pupae, adults and in a Droso-phila cell line (Kc), and stage-specific expression ofseveral different-sized transcripts were recorded. Theresults also suggest that the transcripts found in earlyembryos are of maternal origin since they are foundonly in the ovaries (Madhavan, Bilodeau-Wentworth& Wadsworth, 1985).

(10.3) c-myc expression in teratocarcinoma cellsThe steady-state levels of c-myc found in proliferatingcells have been shown to decrease drastically (Dony,Kessel & Gruss, 1985) in F9 EC cells at a stage evenpreceding overt differentiation induced by retinoicacid 72 h later (Griep & DeLuca, 1986). Post-tran-scriptional mechanisms are apparently wholly respon-sible for reduced c-myc mRNA levels in F9 cells. InF9 cells stimulated to differentiate, the steady state ofc-myc is reduced by as much as 50 % within 3 h (Griep

Page 11: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 459

& DeLuca, 1986). However, if F9 cells are blockedfrom differentiating in response to RA by the ad-dition of 5 mM-sodium butyrate to the medium(Levine, Campisi, Wang & Gudas, 1984) the cells stillrespond to RA by decreased levels of c-myc. Itappears that reduced levels of c-myc are not sufficientfor differentiation but may precede differentiation orreduced growth rates. The undermethylation of thec-myc gene in F9 EC cells relative to differentiatedteratocarcinoma cells and mouse liver DNA mayaccount for its high level of expression and for itsregulation (Griep & DeLuca, 1986). The second exonof the c-myc gene becomes methylated during F9differentiation and this is remarkable in the face ofglobal demethylation of every other gene tested(Young & Tilghman, 1984; Razin et al. 1984).

(11) c-sls

The predicted sequence of p28v~J", the transformingprotein of the simian sarcoma virus (SSV), is strik-ingly homologous to the B chain of PDGF (Water-field et al. 1983; Robbins et al. 1983; Chiu et al. 1984)or PDGF-2 (Rao et al. 1986). In SSV-transformedcells, p28VJ" is proteolytically processed to generate adisulphide-linked dimer (Johnson, Betsholtz, Heldin& Westermark, 1986), and its presence and activity inthe culture medium can be detected and neutralizedby antibodies to PDGF. Using \-sis probes, tran-scripts of c-sis have been detected in many cell lines,tumours and tissues (Eva etal. 1982). Transformationof cells by a wide range of oncogenic agents appearsto activate a cellular gene encoding a PDGF-likemolecule and the induced expression of the c-sis geneby TGF/3 has been suggested as the intermediary stepin the transformed phenotype induced by TGF/3(Leof, Proper, Getz & Moses, 1986). Mouse embryofibroblast cells stimulated with TGF/J express acti-vated the c-sis gene after 4 h with a peak at 12-16 hand release a PDGF-like activity into the culturemedium, c-fos is also induced maximally at 4h withthe appearance of PDGF activity, while c-mycmRNA levels peak at 8-12 h suggesting that PDGFmay have activated both genes. Since TGF/? is pres-ent in most normal tissues (Roberts et al. 1981) andalso in the placenta, serum (Stromberg & Twardzik,1985) and blood platelets (Assoian et al. 1983), it ispossible that fetal growth may be modulated, at leastlocally, by the release of TGF£ and PDGF.

(11.1) c-sis in developmentThe developmental^ regulated expression of the c-sisgene has been mentioned in section 10 since c-mycand c-sis expression occur together in the highlyproliferative cytotrophoblast cells of the first trimes-ter cytotrophoblast shell. What is interesting about

this study (Goustin et al. 1985) is that placentalexplants secrete a PDGF-like activity into the me-dium in a similar developmental time course. Inaddition, cytotrophoblast-like cell lines establishedfrom early placentae display PDGF receptors withsimilar affinity for PDGF as fibroblast cells. Thesecells also respond to PDGF by tenfold elevation ofc-myc mRNA levels in 2 h and by increased synthesisof c-myc protein (several-fold stimulation within 7h).Autocrine stimulation of cytotrophoblast cells by thec-sis product that then results in c-myc and c-fosexpression is an attractive hypothesis. However, theplacenta produces a number of other growth factorssuch as IGFs, FGF and TGF/3, and these could alsocontribute.

PDGF is thought to mediate the proliferation ofsmooth muscle cells in injured arteries and may beinvolved in the pathogenesis of atherosclerosis.Although in injury, the main source of PDGF is theplatelet, other cell types produce PDGF-like activity.For instance, rat aortic smooth muscle cells isolatedfrom 13- to 18-day-old rat pups secrete a PDGFactivity into the medium, and could account forautocrine stimulation of growth of these cells inculture. This activity is developmentally controlledsince the same cell type isolated from adult rats doesnot secrete PDGF (Seifert, Schwartz & Bowen-Pope,1984). c-sis transcripts have been detected at moder-ate levels in cultured human and bovine endothelialcells, at low levels in in vivo endothelium from humanumbilical vein and at very low levels in bovine aorticendothelium in vivo (Barrett et al. 1984). Theseresults also suggest that the sis gene is activated incertain cell types when removed from in vivo to invitro conditions.

Model systems using murine EC cells have sugges-ted that early embryonic stem cells may produce aPDGF-like activity (Gudas, Singh & Stiles, 1983;Rizzino & Bowen-Pope, 1985) but fail to bind thisgrowth factor. It has not been definitively determinedif these cells lack PDGF receptors, or if they arewholly down-regulated by the secreted PDGF. How-ever, when F9, PC13 and PSA1 cells differentiate,they form cell types that are able to bind and respondto PDGF. If the corresponding embryonic cells,namely early embryonic ectoderm cells, producePDGF-like factors, these could then stimulate thegrowth of the adjacent, more differentiated celltypes, such as mesoderm cells which arise on the 7thday of gestation in mouse. PDGF also has a potentchemoattractant activity that stimulates the motilityof certain types of cells and this could play a role inthe migration of embryonic cells. Embryonic ecto-derm cells could also produce other growth factorsshown to be secreted by EC cells (Rizzino, 1982;Heath, Mahadevan & Foulkes, 1986).

Page 12: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

460 E. D. Adamson

(12) Other proto-oncogenes

The latest of the viral oncogenes to be identified witha cellular protein is v-erb-A, which has 89% hom-ology to the thyroid hormone (T3) receptor (Wein-berger et al. 1986). Thyroid hormone is known to beessential for tadpole metamorphosis and the switch toadult-type gene activities (Knowland, 1984). Thisreceptor occurs widely in mammalian tissues butplays important roles in the liver and brain (reviewedby Oppenheimer, 1979). The receptors have higheraffinity for T3 than T4, exist in two molecular forms(Casanova et al. 1984) and appear to be largelynuclear (Erkenbrack & Rosenberg, 1986). Since thy-roid hormone is synthesized throughout gestation,presumably the receptor is also an early product indevelopment.

The c-erb-A gene is related (22 % at the amino acidlevel) to the glucocorticoid receptor (Weinberger etal. 1985; Hollenberg et al. 1985) which is also partlynuclear and partly cytoplasmic. The glucocorticoidreceptor is expressed in many cell types and is activeduring embryogenesis in fetal liver, visceral yolk sac(G. Andrews, personal communication) and the sec-ondary palate (Diewart & Pratt, 1981; Kim, Lauder,Joh & Pratt, 1984).

Two proto-oncogenes originally identified asmouse genomic sequences adjacent to integratedproviruses of the mouse mammary tumour virus(MMTV) in a large proportion of tumours in mice,int-1 and int-2 are unrelated genes, int-2 is transcribedin embryos from day 8-5 to 12-5 and also is seen inadult testis but not other adult tissues (Jakobovits,Shackleford, Varmus & Martin, 1986). This limiteddistribution should be helpful in determining thefunction of the int gene.

(13) Proto-oncogene expression interatocarcinoma cells

Insufficient data have been accumulated to evaluateteratocarcinoma cells as model systems for studyingproto-oncogene mechanisms in growth and differen-tiation, but the potential is great since the stem cellsmay be engineered to express experimentally intro-duced genes. The effect on the stem cell and on thepattern of differentiation produced should be highlyinstructive. The studies of Muller & Wagner (1984)and Ruther et al. (1985) have clearly suggested a rolefor c-fos in differentiation, for example. Once an ECcell line has been established that can express indi-vidual oncogenes, the cells may be aggregated withnormal embryonic cells at the morula stage, ormicroinjected at the blastocyst stage, to follow itseffect on the development of the resulting chimaericanimals. The proportion of engineered cells in each

tissue of the chimaera will vary in individual animalsand may allow a titration of dose versus effect.

To date, Table 2 summarizes what we know of thelevels of oncogene expression in teratocarcinomacells as transcripts that hybridize with DNA probes.As expected, the oncogenes most closely associatedwith proliferation are strongly expressed: these in-clude c-src, c-abl, c-rasHa, c-ras1^, c-myc, and N-myc.When rates of proliferation decline as differentiationoccurs, several oncogenes decline in mRNA level:c-abl and c-ras decline modestly; c-myc and N-mycdecline drastically. Neuronal differentiation is ac-companied by significant increases in c-src (Sejersenet al. 1985; Lynch et al. 1986), but N-myc activitydecreases to low levels (Sejersen et al. 1986). Thenuclear oncogene, c-fos, is expressed transiently atsome point during differentiation that may indicate arole in some general step of that process (Edwards &Adamson, 1986).

Some studies have centred on introducing viral oractivated oncogenes into cell lines to observe effectson cell phenotype and differentiation. For instance,an activated ras gene, c-ras^3, introduced into P19 ECcells appears to have little or no effect and does notprevent the induction of differentiation in response toretinoic acid (Bell, Jardine & McBurney, 1986).However, viruses expressing v-fos (especially FBJ-MSV) are able to immortalize or transform mouseembryo-derived primary cell cultures of fibroblasts,myoblasts, and other cell types (Jenuwein, Muller,Curran & Muller, 1985), thus linking \-fos expressionwith increased growth potential in embryonic cells.Since c-fos can also transform established cell linesunder the right conditions (Miller et al. 1984), thedistinction, if any, between the properties of v-fos andc-fos proteins is not clear. The specific cell typeexpressing the c-fos protein may modulate the out-come. The specific processed forms of oncogeneproducts such as c-fos, c-src and c-erb-A may differamong tissues and may give rise to differences infunction.

(14) Conclusions

For most oncogenes, a specific function or activity hasnot been assigned, and data are still too sparse tohypothesize a mechanism in oncogenesis or to predicta role in development. In the cases of c-erb-B/EGFreceptor, c-fms/CSF-1 receptor, c-sis/PDGF, anobvious niche in growth regulation can be deduced.Although these components are not direct generegulatory molecules, they do have wide-rangingeffects through their interconnections with a family ofprotein kinases and through their effects on phos-phoinositide metabolism, ion fluxes and nutrient

Page 13: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

int-1

int-2

Oncogenes in development 461

Table 2. Expression o/c-oncs in teratocarcinoma cells

c-onc

c-erb-B/EGF-R

c-fms

c-src

c-mosc-fesc-mybc-erb-A

c-abl

c-ras

c-myc

N-myc

c-fos

c-sis/PDGF-like factor

CeU line

F9, PC13OC15

P19

HT-H

PCC7

F9P1951801A1

PC13, F9, 3TDMPYS-2, PSA5E, C110

PC13, F9, 3TDMPYS-2, PSA5E, C110

OC15, F9, HR9

PC13, F9, 3TDMPYS-2, PSA5E, C110

PCC4

PC13, F9, 3TDMPYS-2, PSA5E, C110

F9

human ECCmouse ECC

PCC7, PCC3,PCC4, F9

F9, OC15

F9

F9

P19

PCC4

PCC3 etc.F9, PC13

Exp" in ECC

-Intracellular

autophos. acty

-

-

+

-+

-

low

++ +

+ + + geneamplified

+

+ +

+ +

+ +

low

low

low

low

+

lowlow

Exp" indifferentiated cells

++ +

+

trophoblast-likecells +

3—5x inc. duringneuronal diff '

-+ + +

change in mobility

-

low

++

lower

-

Post-transcriptionaldown-regulation within

24 h of inductionof differentiation

85 % reduction duringnerve differentiation

low

5x inc. by day 3 withRA. Aggregates

No inc. with RA &cAMP. Monolayers

20 x inc. by day 12 withDMSO. Aggregates

lower

low-

Reference

Reese/ al. 1979;Adamson & Hogan, 1984;Welter er al. 1987.

Mummery er al. 1985.

Izhar er al. 1986.

Sejersen er al. 1985.

Sejersen er al. 1985.

Lynch er al. 1986.

Sejersen ex al. 1985.

Sejersen er al. 1985.

Muller, 1983.

Sejersen er al. 1985.

Vilette er al. 1985.

Sejersen er al. 1985.

Dony era/. 1985;Sejersen er al. 1986.

Jakobovitz er al. 1985.

Sejersen er al. 1986.

Muller, 1983.

Edwards & Adamson(unpublished data)

Mason er al. 1985.

Edwards & Adamson,1986.

Vilette era/. 1985.

Sejersen er al. 1985.Gudas er al. 1983;Rizzino & Bowen-Pope,1985.

F9, PSA

F9, PSA

Jakobovits ef al. 1986.

low

Footnotes: RA = retinoic acid; cAMP = dibutyryl cyclic AMP; DMSO = dimethyl sulphoxide.

transport. Nevertheless, the role of these growth-related proteins is poorly understood. Largely this isdue to their complicated interconnections with manycellular metabolic processes. Indeed, with a fewexceptions such as PDGF, the oncogenes seem to belargely concerned with general metabolic processes as

much as (if not more than) growth regulation per se.The proto-oncogenes that are known to have enzy-matic activity, such as c-src, c-abl, the ras family, alsofit into this metabolic framework. c-erb-A/thyroidhormone receptor is even more directly concernedwith overall metabolic rates.

Page 14: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

462 E. D. Adamson

The nuclear-located proto-oncogenes are betterplaced for roles in growth/differentiation/gene regu-lation. They bind to DNA to various degrees,although specific binding sites have not been ident-ified. The c-fos protein appears to bind to chromatin,particularly at deoxyribonuclease I-sensitive sites,thus indicating a role in the regulation of geneexpression (Sambucetti & Curran, 1986; Renz,Verrier, Kurz & Muller, 1987). c-myc production issimilarly activated but usually at a slightly later time.This has given rise to the possibility of chains ofcommand leading to gene-stimulating events gener-ated by signals from the environment, c-fos and c-mycmay then activate sets of genes that lead to DNAsynthesis and progression through the cell cycle, or tosets of genes that regulate differential cell-lineagepathways.

TGF0

IPDGF-—PDGF-R—-c-fos-"C-myc—Tas •*.

TPA- pK-C—*-c-fos-~-c-myc-~-c-fms

EGF -EGF-R—-c-fos-^c-myc—»• J

proliferation

differentiation

TGFa- ras

In reviewing the data here it becomes clear thatalmost without exception, the proto-oncogenes havebeen shown to play roles in differentiation as wellas proliferation. Is this because directing the celltowards proliferation limits the cell's resources tocontinue the previous commitment or directiontowards a differentiated phenotype (e.g. c-mycl). Oris it a more direct process that facilitates a differentia-tive pathway (e.g. c-fos?). Only in one case is thefunction clear, that is, c-fms and this product appearslate and may only be a marker and not a regulator ofdifferentiation.

For considerations of the roles of proto-oncogenesin development, some specificity of action may residein c-src since a neurone-specific isotype has beenidentified in brain during proliferation, commitmentand expression of the neuronal phenotype. Otherproto-oncogenes are less well defined to specifictissues. The placenta and other extraembryonic tis-sues (VYS, amnion, and chorion), however, do holda special place in the high levels of expression ofseveral c-oncs. In addition to c-myc, c-abl, c-ras, andc-src, very high levels of c-sis, c-fos, and c-fms occur,and these levels vary between the tissues mentionedand between the temporal stages of the gestational

period. This makes it less likely that the undermeth-ylated state of the DNA in these tissues (Rossant,Sanford, Chapman & Andrews, 1986) broadly dic-tates the levels of proto-oncogene expression. It isstill possible that specific sites of methylated basescontrol the expression of these or other genes in atemporal and tissue-specific manner. The undermeth-ylated state of the placenta could be necessary for thederepression of a wide range of growth-related genesthat are known to be active in the placenta. In thepresence of secreted 'autocrine' growth factors suchas TGFa", TGF0, PDGF, IGF-I, and IGF-II, recep-tors are continually down-regulated, synthesized andactivating the chain of command. Although thesetissues have been recognized as 'pseudomalignant,'they are not tumorigenic in nude mice and havesomehow controlled the signal to proliferate in paral-lel to the needs of the growing fetus. Studies onmodes of oncogene activation in carcinogenesis showthat a quantitative change of expression is one mech-anism of such activation (reviewed by Klein & Klein,1984). Understanding the mechanisms of control inthe placenta may therefore be instructive for futureclinical applications (Conway, 1983).

Limitations of space did not allow recognition of allrelevant references; for these omissions my apologies areoffered. I am grateful to Drs C. Van Beveren, C. Der, andS. Edwards for comments on the manuscript. This work wassupported by grants CA 28424, P30 CA 30199 and HD21957 from the National Institutes of Health.

References

ADAMS, J. M., HARRIS, A. W., PINKERT, C. A.,CORCORAN, L. M., ALEXANDER, W. S., CORY, S.,PALMITER, R. D. & BRINSTER, R. L. (1985). The c-mycgene driven by immunoglobulin enhancer induceslymphoid malignancy in transgenic mice. Nature, Lond.318, 533-538.

ADAMSON, E. D. (1983). Growth factors in development.In The Biological Basis of Reproductive andDevelopmental Medicine (ed. J.B. Warshaw), pp.307-336. New York: Elsevier Press.

ADAMSON, E. D. (1986). Cell lineage-specific geneexpression in development. In Experimental Approachesto Mammalian Embryonic Development (ed. J. Rossant& R. Pedersen). New York: Cambridge UniversityPress (in press).

ADAMSON, E. D., DELLER, M. J. & WARSHAW, J. B.(1981). Functional EGF receptors on embryonic cells.Nature, Lond. 291, 656-659.

ADAMSON, E. D. & HOGAN, B. L. M. (1984). Expressionof EGF receptor and transferrin by F9 and PCDteratocarcinoma cells. Differentiation 27, 152-157.

ADAMSON, E. D. & MEEK, J. (1984). The ontogeny ofepidermal growth factor receptors during mousedevelopment. Devi Biol. 103, 62-71.

Page 15: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 463

ADAMSON, E. D., MEEK, J. & EDWARDS, S. A. (1985).Product of the cellular oncogene, c-fos, observed inmouse and human tissues using an antibody to asynthetic peptide. EMBO J. 4, 941-947.

ADAMSON, E. D. & REES, A. R. (1981). Epidermalgrowth factor receptors. Mol. Cell. Biochem. 34,129-152.

ADAMSON, E. D. & WARSHAW, J. B. (1981). "Down-regulation" of epidermal growth factor receptors inmouse embryos. Devi Biol. 90, 430-434.

ALEMA, S., CASALBORE, P., AGOSTINI, E. & TATO, F.(1985). Differentiation of PC12 pheochromocytoma cellsinduced by v-src oncogene. Nature, Lond. 316, 557-559.

ALEMA, S., TATO, F. & BOETTIGER, D. (1985). myc and src

oncogenes have complementary effects on cellproliferation and expression of specific extracellularmatrix components in definitive chondroblasts. Mol.Cell. Biol. 5, 538-544.

ALTTALO, K., RAMSEY, G., BISHOP, J. M., PFEIFER, S. O.,COLBY, W. W. & LEVINSON, A. D. (1983).Identification of nuclear proteins encoded by viral andcellular myc oncogenes. Nature, Lond. 306, 274-277.

ANDREWS, G. K., FIARDING, M. A., CALBET, J. P. &ADAMSON, E. D. (1987). Elevated expression of c-fosduring heat shock of FfeLa cells (submitted).

ASSOIAN, R. K., KOMORTYA, A., MEYERS, C. A., MILLER,D. M. & SPORN, M. B. (1983). Transforming growthfactor-/3 in human platelets: Identification of a majorstorage site, purification, and characterization. / . biol.Chem. 258, 7155-7160.

BALDWIN, G. S. (1985). Epidermal growth factorprecursor is related to the translation product of theMoloney sarcoma virus oncogene mos. Proc. natn.Acad. Sci. U.S.A. 82, 1921-1925.

BARBACID, M., LAUVER, A. V. & DEVARE, S. G. (1980).Biochemical and immunological characterization ofpolyproteins coded for by the McDonough GardnerArnstein and Snyder-Theilen strains of feline sarcomavirus. / . Virol. 33, 196-207.

BARKA, T., GUBITS, R. M. & VANDER NOEN, H. (1986).Beta-adrenergic stimulation of c-fos gene expression inthe mouse submandibular gland. Mol. Cell. Biol. 6,2984-2989.

BARRETT, T. B., GAJDUSEK, C. M., SCHWARTZ, S. M.,MCDOUGALL, J. K. & BENDITT, E. P. (1984).Expression of the sis gene by endothelial cells inculture and in vivo. Proc. natn. Acad. Sci. U.S.A. 81,6772-6774.

BAR-SAGI, D. & FERAMISCO, J. R. (1985). Microinjectionof the ray oncogene protein into PC12 cells inducesmorphological differentiation. Cell 42, 841-848.

BELL, J. C , JARDINE, K. & MCBURNEY, M. W. (1986).Lineage-specific transformation after differentiation ofmulti-potential murine stem cells containing a humanoncogene. Mol. Cell. Biol. 6, 617-625.

BESMER, P., MURPHY, J. E., GEORGE, P. C , Qru, F.,BERGOLD, P. J., LEDERMAN, L., SNYDER, H. W.,BRODEUR, D., ZUCKERMAN, E. E. & HARDY, W. D.(1986). A new acute transforming feline retrovirus andrelationship of its oncogene v-kit with the proteinkinase gene family. Nature, Lond. 320, 415-422.

BIRNIE, G. D., BURNS, J. H., CLARK, P. & WARNOCK, A.M. (1984). Lineage-specific and differentiation-stage-specific gene expression in normal and leukemic humanmyeloid cells. J. Roy. Soc. Med. 77, 289-294.

BISHOP, J. M. (1985). Viral oncogenes. Cell 42, 23-38.BLANCHARD, J.-M., PIECHACZYK, M., DANI, C ,

CHAMBARD, J.-C, FRACHI, A., POUYSSEGUR, J. &JEANTEUR, P. (1985). C-myc is transcribed at high ratein Go-arrested fibroblasts and is post-transcriptionallyregulated in response to growth factors. Nature, Lond.317, 443-445.

BOETTIGER, D., ANDERSON, S. & DEXTER, T. M. (1984).Effect of src infection on long term marrow cultures:Increased self-renewal of hemopoietic progenitor cellswithout leukemia. Cell 36, 763-773.

BRAVO, R., BURCKHARDT, J., CURRAN, T. & MULLER, R.(1985). Stimulation and inhibition of growth by EGF indifferent A431 cell clones is accompanied by rapidinduction of c-fos and c-myc proto-oncogenes. EMBOJ. 4, 1193-1197.

BRUGGE, J. S., COTTON, P. C , QUERAL, A. E., BARRETT,J. N., NUNNER, D. & KEANE, R. W. (1985). Neuronesexpress high levels of a structurally modified activatedform of ppflT"*. Nature, Lond. 316, 554-557.

CAMPISI, J., GRAY, H. E., PARDEE, A. B., DEAN, M. &SONENSHEIN, G. E. (1984). Cell-cycle control of c-mycbut not c-ras expression is lost following chemicaltransformation. Cell 36, 241-247.

CARLIN, C. R. & ANDREWS, P. W. (1985). Humanembryonal carcinoma cells express low levels offunctional receptor for epidermal growth factor. ExplCell Res. 159, 17-26.

CARMIER, J. F. & SAMARUT, J. (1980). Chicken myeloidstem cells infected by retroviruses carrying the v-fpsoncogene do not require exogenous growth factors todifferentiate in vitro. Cell 44, 159-165.

CARPENTER, G. (1984). Properties of the receptor forepidermal growth factor. Cell 37, 357-358.

CARPENTER, G. & ZENDOQUI, J. G. (1986). Epidermalgrowth factor, its receptor and related proteins. ExplCell Res. 164, 1-10.

CASANOVA, J., HOROWITZ, Z. D., COPP, R. P., MCINTYRE,

W. R., PASCUAL, A. & SAMUELS, H. H. (1984).Photoaffinity labeling of-thyroid hormone nuclearreceptors. / . biol. Chem. 259, 12084-12091.

CATTERTON, W. Z., ESCOBEDO, M. B. & SEXSON, W. R.

(1979). Effect of EGF on lung maturation in fetalrabbits. Ped. Res. 13, 104-108.

CHANG, C.-C, TROSKO, J. E., KUNG, H.-J., BOMBICK, D.& MATSUMURA, F. (1985). Potential role of the src geneproduct in inhibition of gap-junctional communicationin NLH/3T3 cells. Proc. natn. Acad. Sci. U.S.A. 82,5360-5364.

CHIU, I.-M., REDDY, E. P., GIVOL, D., ROBBINS, K. C ,TRONICK, S. R. & AARONSON, S. A. (1984). Nucleotidesequence analysis identifies the human c-sis proto-oncogene as a structural gene for platelet-derivedgrowth factor. Cell 37, 123-129.

COHEN, S. (1962). Isolation of a mouse submaxillarygland protein accelerating incisor eruption and eyelid

Page 16: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

464 E. D. Adamson

opening in the new born animal. / . biol. Chem. 237,1555-1562.

COLLETT, M. S. & ERIKSON, R. L. (1978). Protein kinaseactivity associated with the avian sarcoma virus srcgene product. Proc. natn. Acad. Sci. U.S.A. 75,2021-2024.

COLLETTA, G., CIRAFICI, A. M. & VECCHIO, G. (1986).Induction of the c-fos oncogene by thyrotropichormone in rat thyroid cells in culture. Science 233,458-460.

CONSCIENCE, J.-F., VERRIER, B. & MARTIN, G. (1986).Interleukin-3-dependent expression of the c-myc and c-fos proto-oncogenes in hemopoietic cell lines. EMBOJ. 5, 317-323.

CONTEAS, C. N., DEMORROW, J. M. & MAJUMDAR, A. P.N. (1986). Effect of epidermal growth factor on growthand maturation of fetal and neonatal rat small intestinein organ culture. Experientia 42, 950-952.

CONWAY, F. J. (1983). A proposed model of cancer as theinappropriate expression of non-body introns. / . theor.Biol. 100, 1-24.

COOPER, C. S., BLAIR, D. G., OSKARSSON, M. K.,TAINSKY, M. A., EADER, L. A. & VANDE WOUDE, G.F. (1984). Characterization of human transforminggenes from chemically transformed, teratocarcinoma,and pancreatic carcinoma cell lines. Cancer Res. 44,1-10.

COTTON, P. C. & BRUGGE, J. S. (1983). Neural tissuesexpress high levels of the cellular src gene productp p ^ ^ . Mol. Cell Biol. 3, 1157-1162.

COUSSENS, L., VAN BEVEREN, C , SMITH, D., CHEN, E.,MITCHELL, R. L., ISACKE, C. M., VERMA, I. M. &ULLRICH, A. (1986). Structural alteration of viralhomologue of receptor proto-oncogene fms at carboxyterminus. Nature, Lond. 320, 277-280.

COWLEY, G., SMITH, J. A., GUSTERSON, B., HENDLER, F.& OZANNE, B. (1984). The amount of epidermalgrowth factor receptor is elevated in squamous cellcarcinomas. Cancer Cells 1, 5-10.

CURRAN, T., MILLER, A. D., ZOKAS, L. & VERMA, I. M.(1984). Viral and cellular fos proteins: A comparativeanalysis. Cell 36, 259-268.

CURRAN, T. & MORGAN, J. I. (1985). Superinduction of c-fos by nerve growth factor in the presence ofperipherally active benzodiazepines. Science 229,1265-1268.

CURRAN, T. & TEICH, N. M. (1982). Candidate product ofthe FBJ murine osteosarcoma virus oncogene:characterization of a 55000 dalton phosphoprotein. / .Virol. 42, 114-122.

CURRAN, T. & VERMA, I. M. (1984). FBR Murineosteosarcoma virus: I. Molecular analysis andcharacterization of a 75,000 Da gag-/os fusion product.Virology 135, 218-228.

DEAN, M., LEVTNE, R. A. & CAMPISI, J. (1986). c-mycregulation during retinoic acid-induced differentiationof F9 cells is post-transcriptional and associated withgrowth arrest. Mol. Cell. Biol. 6, 518-524.

DECINO, P., HERBST, H., LERNHARDT, W. & RASCHKE,W. C. (1987). Expression of oncogenes in normal andtransformed murine B lymphocytes (submitted).

DEFEO, D., GONDA, M. A., YOUNG, H. A., CHANG, E.H., LOWY, D. R., SCOLNICK, E. M. & ELLIS, R. W.(1981). Analysis of two divergent rat genomic cloneshomologous to the transforming gene of Harveymurine sarcoma virus. Proc. natn. Acad. Sci. U.S.A. 78,3328-3332.

DEKEL, N. & SHERIZLY, I. (1985). EGF inducesmaturation of rat follicle-enclosed oocytes.Endocrinology 116, 406-409.

DEMBINSH, A. B. & JOHNSON, L. R. (1985). Effect ofEGF on the development of rat gastric mucosa.Endocrinology 116, 90-94.

DERYNCK, R., ROBERTS, A. B., WINKLER, M. E., CHEN,E. Y. & GOEDDEL, D. V. (1984). Human transforminggrowth factor-a-: precursor structure and expression inE. coli. Cell 38, 287-297.

DESCHAMPS, J., MITCHELL, R. L., MEJLINK, F., KRUJIER,W., SCHUBERT, D. & VERMA, I. M. (1985). Proto-oncogene fos is expressed during development,differentiation and growth. Cold Spring Harb. Symp.quant. Biol. 50, 733-745.

DEVARE, S. G., REDDY, E. P., LAW, J. D., ROBBINS, K.C. & AARONSON, S. (1983). Nucleotide sequence of thesimian sarcoma virus genome: demonstration that itsacquired cellular sequences encode the transforminggene product p28J". Proc. natn. Acad. Sci. U.S.A. 80,731-735.

DHAR, R., ELLIS, R. W., SHIH, T. Y., OROSZIAN, S.,SHAPIRO, B., MAIZEL, J., LOWY, D. & SCOLNICK, E.(1982). Nucleotide sequence of the p21 transformingprotein of Harvey murine sarcoma virus. Science 217,934-937.

DIEWART, V. M. & PRATT, R. M. (1981). Cortisone-induced cleft palate in the A/J mouse: failure of palatalself contact. Teratology 24, 149-153.

DONY, C , KESSEL, M. & GRUSS, P. (1985). Post-transcriptional control of myc and p53 expressionduring differentiation of the embryonal carcinoma cellline F9. Nature, Lond. 317, 636-639.

DOOUTTLE, R. F., HUNKAPILLER, M. W., HOOD, L. E.,DEVARE, S. G., ROBBINS, K. C , AARONSON, S. A. &ANTONIADES, H. N. (1983). Simian sarcoma virusoncogene, \-sis, is derived from the gene (or genes)encoding a platelet-derived growth factor. Science 221,275-276.

DOWNWARD, J., YARDEN, Y., MAYES, E., SCRACE, G.,TOTTY, N., STOCKWELL, P., ULLRICH, A., SCHLESSINGER,J. & WATERFIELD, M. D. (1984). Close similarity ofepidermal growth factor receptor and \-erb-B oncogeneprotein sequences. Nature, Lond. 307, 521-527.

DUPREY, S. P. & BOETTIGER, D. (1985). Developmentalregulation of c-myb in normal myeloid progenitor cells.Proc. natn. Acad. Sci. U.S.A. 82, 6937-6941.

EDWARDS, S. A. & ADAMSON, E. D. (1986). Induction ofc-fos and AFP expression in differentiating embryonalcarcinoma cells. Expl Cell Res. 165, 473-480.

EISENMANN, R. N., TACHTBANA, C. Y., ABRAMS, H. D. &HANN, S. R. (1985). v-Myc and c-myc-encoded proteinsare associated with the nuclear matrix. Mol. Cell. Biol.5, 114-126.

Page 17: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 465

ELLIS, R. W., DEFEO, D., FURTH, M. E. & SCOLNICK, E.M. (1982). Mouse cells contain two distinct ras genemRNA species that can be translated into a p21 oneprotein. Mol. Cell. Biol. 2, 1335-1345.

EMILIA, G., DONELLI, A., FERRARI, S., TORELLI, U.,SELLERI, L., ZUCCHINI, P., MORETTI, L., VENTURELU,D., CECCHERELU, G. & TORELU, G. (1986). Cellularlevels of mRNA from c-myc, c-myb and c-fos one-genesin normal myeloid and erythroid precursor of humanbone marrow: an in situ hybridization study. Brit. J.Haematol. 62, 287-292.

ERICKSON, C. A. & TURLEY, E. A. (1987). The effects ofepidermal growth factor on neural crest cells in tissueculture. Expl Cell Res. (in press).

ERKENBRACK, D. E. & ROSENBERG, L. L. (1986). Bindingof thyroid hormone to nuclear extracts of thyroid cells.Endocrinology 119, 311-317.

EVA, A., ROBBINS, K. C , ANDERSEN, P. R., SRINIVASAN,A., TRONICK, S. R., REDDY, E. P., ELLMORE, N. W.,GALEN, A. T., LAUTENBERGER, J. A., PAPAS, T. S.,WESTIN, E. H., WONG-STAAL, F., GALLO, R. C. &AARONSON, S. A. (1982). Cellular genes analogous toretroviral one genes are transcribed in human tumourcells. Nature, Lond. 295, 116-119.

FERRARI, S., TORELLI, U., SELLERI, L., CONELLJ, A.,VENTURELU, D., MORETTI, L. & TORELLI, G. (1985).Expression of human c-fes one-gene occurs atdetectable levels in myeloid but not in lymphoid cellpopulations. Brit. J. Haematol. 59, 21-25.

FINIDORI-LEPICARD, J., SCHORDERET-SLATKINE, S.,HANOUNE, J. & BAULIEU, E. E. (1981). Progesteroneinhibits membrane bound adenylate cyclase in Xenopuslaevis oocytes. Nature, Lond. 292, 255-257.

FINKEL, T., DER, C. J. & COOPER, G. M. (1984).Activation of ras genes in human tumors does notaffect localization, modification or nucleotide bindingproperties of p21. Cell 37, 151-158.

FLEISCHMAN, L. F., CHAHWALA, S. B. & CANTLEY, L.(1986). /fas-transformed cells: altered levels ofphosphatidylinositol-4,5 biphosphate and catabolites.Science 231, 407-410.

FULTS, D. W., TOWLE, A. C , LAUDER, J. M. & MANESS,P. F. (1985). ppW"^ in the developing cerebellum.Mol. Cell. Biol. 5, 27-32.

GEBHARDT, A., BELL, J. C. & FOULKES, J. G. (1986).Abelson transformed fibroblasts lacking the EGFreceptor are not tumourigenic in nude mice. EMBO J.5, 2191-2195.

GESSLER, M. & BARNEKOW, A. (1984). Differentialexpression of the cellular oncogenes c-src and c-yes inembryonal and adult chicken tissues. Bioscience Reports4, 757-770.

GILMAN, A. G. (1984). G proteins and dual control ofadenylate cyclase. Cell 36, 577-579.

GOFF, S. P., GlLBOA, E., WlTTE, O. N. & BALTIMORE, D.(1980). Structure of the Abelson murine leukemia virusand the homologous cellular gene: studies with clonedviral DNA. Cell 22, 777-783.

GOLDIN, G. V. & OPPERMAN, L. A. (1980). Induction ofsupernumerary tracheal buds and the stimulation of

DNA synthesis in the embryonic chick lung andtrachea by EGF. /. Embryol. exp. Morph. 60, 235-247.

GONDA, T. J., GOUGH, N. M., DUN, A. R. &DEBLAQUIERE, J. (1985). Nucleotide sequence of cDNAclones of the murine myb proto-oncogene. EMBO J. 4,2003-2008.

GONDA, T. J. & METCALF, D. (1984). Expression oimyb,myc and fos proto-oncogenes during the differentiationof a murine myeloid leukemia. Nature, Lond. 310,249-251.

GOUSTIN, A. S., BETSHOLTZ, C , PPEIFER-OHLSSON, S.,PERSSON, H., RYDNERT, J., BYWATER, M., HOLMGREN,G., HELDIN, C.-H., WESTERMARK, B. & OHLSSON, R.(1985). Coexpression of the sis and myc proto-oncogenes in developing human placenta suggestsautocrine control of trophoblast growth. Cell 41,301-312.

GOYETTE, M . , PETROPOULOS, C. J . , SHANK, P . R. &FAUSTO, N. (1984). Regulated transcription of c-Ki-rasand c-myc during compensatory growth of rat liver.Mol. Cell. Biol. 4, 1493-1498.

GREENBERG, M. E., GREENE, L. A. & ZIFF, E. B. (1985).Nerve growth factor and epidermal growth factorinduce rapid transient changes in proto-oncogenetranscription in PC12 cells. J. biol. Chem. 260,14101-14110.

GREENBERG, M. E., HERMANOWSKI, A. L. & ZIFF, E. B.(1986). Effect of protein synthesis inhibitors on growthfactor activation of c-fos, c-myc, and actin genetranscription. Mol. Cell. Biol. 6, 1050-1057.

GREENBERG, M. E. & ZIFF, E. B. (1984). Stimulation of3T3 cells induces transcription of the c-fos proto-oncogene. Nature, Lond. 311, 433-437.

GRIEP, A. E. & DELUCA, H. F. (1986). Decreased c-mycexpression is an early event in retinoic acid-induceddifferentiation of F9 teratocarcinoma cells. Proc. natn.Acad. Sci. U.S.A. 87, 5539-5543.

GROSS, J., DYNIA, D. W., ROONEY, S. A., SMART, D. A.,WARSHAW, J. B., SISSOM, J. F. & HOATH, S. B. (1986).Influence of epidermal growth factor on fetal rat lungdevelopment in vitro. Fed. Res. 20, 473-477.

GUBITS, R. M., SHAW, P. A., GRESIK, E. W., ONETTI-MUDA, A. & BARKA, T. (1986). Epidermal growthfactor gene expression is regulated differently in mousekidney and submandibular gland. Endocrinology 119,1382-1387.

GUDAS, L. J., SINGH, J. P. & STILES, C. D. (1983).Secretion of growth regulatory molecules byteratocarcinoma stem cells. Cold Spring Harb. Conf.Cell Prolif. 10, 229-236.

HAGAG, N., HALEGOUA, S. & VIOLA, M. (1986). Inhibitionof growth factor-induced differentiation of PC12 cellsby microinjection of antibody to ras p21. Nature, Lond.319, 680-682.

HALL, A., MARSHALL, C. J., SPURR, N. K. & WEISS, R.A. (1983). Identification of transforming gene in twohuman sarcoma cell lines as a new member of the rasgene family located on chromosome 1. Nature, Lond.303, 396-400.

HAMPE, A., LAPREVOTTE, I., GALIBERT, F., FEDELE, L. A.& SHERR, C. J. (1982). Nucleotide sequences of feline

Page 18: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

466 E. D. Adamson

retroviral oncogenes (v-/es) provide evidence for afamily of tyrosine-specific protein kinase genes. Cell 30,775-785.

HAYMAN, M. J., RAMSAY, G. M., SAVIN, K., KITCHENER,G., GRAF, T. & BEUG, H. (1983). Identification andcharacterization of the avian erythroblastosis virus erbBgene product as a membrane glycoprotein. Cell 32,579-588.

HEATH, J. K., MAHADEVAN, L. & FOULKES, J. G. (1986).The role of EGF receptor transmodulation inembryonal carcinoma-derived growth factor-inducedmitogenesis. EMBO J. 5, 1809-1814.

HELDIN, C.-H. & WESTERMARX, B. (1984). Growthfactors: mechanism of action and relation tooncogenes. Cell 37, 9-20.

HERSCHMAN, H. R. (1985). The EGF receptor. In Controlof Animal Cell Proliferation (ed. A. L. Boynton & H.L. Leffert), pp. 169-200. New York: Academic Press.

HOATH, S. B. (1986). Treatment of the neonatal rat withepidermal growth factor and differences in time andorgan response. Fed. Res. 20, 468-472.

HOLLENBERG, S. M., WEINBERGER, C , ONG, E. S.,CERELLI, G., ORO, A., LEBO, R., THOMPSON, E. B.,ROSENFELD, M. G. & EVANS, R. M. (1985). Primarystructure and expression of a functional humanglucocorticoid receptor cDNA. Nature, Lond. 318,635-642.

HOLT, J. T., GOPAL, T. V., MOULTON, A. D. & NIENHIUS,A. W. (1986). Inducible production oic-fos antisenseRNA inhibits 3T3 cell proliferation. Proc. natn. Acad.Sci. U.S.A. 83, 4794-4798.

HORTSCH, M . , SCHLESSINGER, J . , GOOTWINE, E . & WEBB,C. G. (1983). Appearance of functional EGF receptorkinase during rodent embryogenesis. EMBO J. 2,1937-1941.

HULEIHEL, M . , GOLDSBOROUGH, M . , CLEVELAND, J . ,GUNNELL, M . , BONNER, T . & LAPP, U. R. (1986).Characterization of murine A-raf, a new oncogenerelated to the v-ra/7 oncogene. Mol. Cell. Biol. 6,2655-2662.

HUNG, M.-C, SCHECHTER, A. L., CHEVRAY, P. Y., STERN,D. F. & WEINBERG, R. A. (1986). Molecular cloning ofthe neu gene: Absence of gross structural alteration inoncogenic alleles. Proc. natn. Acad. Sci. U.S.A. 83,261-264.

HUNTER, T. (1984). Oncogenes and proto-oncogenes:How do they differ? J. natn. Cancer Inst. 73, 773-786.

HURLEY, J. B., SIMON, M. I., TEPLOW, D. B., ROBISHAW,J. D. & GILMAN, A. G. (1984). Homologies betweensignal transducing G proteins and ras gene products.Science 226, 860-867.

IZHAR, M . , SlEBERT, P . D . , OSHIMA, R. G. , DE WOLF, W.C. & FUKUDA, M. N. (1986). Trophoblasticdifferentiation of human teratocarcinoma cell line HT-H. Devi Biol. 116, 510-518.

JAKOBOVTTS, A., SCHWAB, M., BISHOP, J. M. & MARTIN,

G. R. (1985). Expression of N-myc in teratocarcinomastem cells and mouse embryos. Nature, Lond. 318,188-191.

JAKOBOVTTS, A., SHACKLEFORD, G. M., VARMUS, H. E. &MARTIN, G. R. (1986). Two proto-oncogenes

implicated in mammary carcinogenesis, int-1 and int-2,are independently regulated during mousedevelopment. Proc. natn. Acad. Sci. U.S.A. 83,7806-7810.

JANSSEN, J. W., VERNOLE, P., DEBOER, P. A.,

OOSTERHUIS, J. W. & COLLARD, J. G. (1986).Sublocalization of c-myb to 6q.21-q23 by in situhybridization and c-myb expression in a humanteratocarcinoma with 6q rearrangements. Cytogenet.Cell Genet. 41, 129-135.

JENUWEIN, T., MOLLER, D., CURRAN, T. & MULLER, R.(1985). Extended life span and tumorigenicity ofnonestablished mouse connective tissue cellstransformed by the fos oncogene of FBR-MuSV. Cell41, 629-637.

JOHNSON, A., BETSHOLTZ, C , HELDIN, C.-H. &WESTERMARK, B. (1986). The phenotypic characteristicsof simian sarcoma virus - transformed humanfibroblasts suggest that the s-sis gene product actssolely as a PDGF receptor agonist in celltransformation. EMBO J. 5, 1535-1541.

KACZMAREK, L. (1986). Protooncogene expression duringthe cell cycle. Lab. Invest. 54, 365-376.

KACZMAREK, L., HYLAND, J. K., WATT, R., ROSENBERG,M. & BASERGA, R. (1985). Microinjected c-myc as acompetence factor. Science 228, 1313-1315.

KAMPS, M. P., BUSS, J. E. & SEFTON, B. M. (1985).Mutation of NH2-terminal glycine of P60"1 preventsboth myristylation and morphological transformation.Proc. natn. Acad. Sci. U.S.A. 82, 4625^628.

KELLY, K., COCHRAN, B. H., STILES, C. D. & LEDER, P.(1983). Cell-specific regulation of the c-myc gene bylymphocyte mitogens and platelet-derived growthfactor. Cell 35, 603-610.

KIM, C. S., LAUDER, J. M., JOH, T. H. & PRATT, R. M.(1984). Immunocytochemical localization ofglucocorticoid receptors in midgestation murineembryos and human embryonic cultured cells. J.Histochem. Cytochem. 32, 1234-1237.

KLEIN, G. & KLEIN, E. C. (1984). Oncogene activationand tumour progression. Carcinogenesis 5, 429-435.

KLEIN, G. & KLEIN, E. (1985). Evolution of tumours andthe impact of molecular oncology. Nature, Lond. 315,190-195.

KNOWLAND, J. S. (1984). Hormones and animaldevelopment. In Developmental Control in Animals andPlants (ed. C. F. Graham & P. F. Wareing), 2ndedition, pp. 349-365. Oxford: Blackwell.

KRETSINGER, R. H. & CREUTZ, C. E. (1986). Consensus inexocytosis. Nature, Lond. 320, 573.

KRIS, R. M., LIBERMANN, T. A., Avrvi, A. &SCHLESSINGER, J. (1985). Growth factors, growth factorreceptors and oncogenes. Biotechnology 3, 135-140.

KRUUER, W., COOPER, J. A., HUNTER, T. & VERMA, I. M.(1984). PDGF induces rapid but transient expression ofthe c-fos gene. Nature, Lond. 312, 711-716.

KRUUER, W., SCHUBERT, D. & VERMA, I. M. (1985).Induction of the proto-oncogene fos by nerve growthfactor. Proc. natn. Acad. Sci. U.S.A. 82, 7330-7334.

KRUUER, W., SKELLY, H., BATTERI, F., VAN DER PUTTEN,H., BARBER, J. R., VERMA, I. M. & LEFFERT, H. L.

Page 19: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 467

(1986). Proto-oncogene expression in regenerating liveris simulated in cultures of primary adult rathepatocytes. J. biol. Chem. 261, 7929-7933.

LACHMAN, H. M. & SKOULTCHI, A. I. (1984). Expressionof c-myc changes during differentiation of mouseerythroleukemia cells. Nature, Lond. 310, 592-594.

LEDER, A., PATTENGALE, P. K., Kuo, A., STEWART, T. A.& LEDER, P. (1986). Consequences of widespreadderegulation of the c-myc gene in transgenic mice:multiple neoplasms and normal development. Cell 45,485-495.

LEE, D. C , ROCHFORD, R., TODARO, G. J. & VILLARREAL,L. P. (1985a). Developmental expression of rattransforming growth factor arnRNA. Mol. Cell Biol. 5,3644-3646.

LEE, D. C , ROSE, T. M., WEBB, N. R. & TODARO, G. J.(1985b). Cloning and sequence analysis of a cDNA forrat transforming growth factor-a-. Nature, Lond. 313,489-492.

LEOF, E. D., PROPER, J. A., GETZ, M. J. & MOSES, H. L.(1986). Transforming growth factor type /3 regulation ofactin mRNA. J. cell. Physiol. 127, 83-88.

LEV, Z., KIMCHIE, Z., HESSEL, R. & SEGEV, D. (1985).Expression of ras cellular oncogene duringdevelopment of Drosophila melanogaster. Mol. Cell.Biol. 5, 1540-1542.

LEVINE, R. A., CAMPISI, J., WANG, S.-Y. & GUDAS, L. J.(1984). Butyrate inhibits the retinoic acid induceddifferentiation of F9 teratocarcinoma stem cells. DeviBiol. 105, 443-450.

LEVY, B. T., SORGE, L. K., MEYMANDI, A. & MANESS, P.F. (1984). pp60c"jrc kinase is in chicken and humanembryonic tissues. Devi Biol. 104, 9-17.

LIBERMANN, T. A., NUSBAUM, H. R., RAZON, N., KRIS,R., LAX, I., SOREQ, H., WHITTLE, N., WATERFIELD, M.D., ULLRICH, A. & SCHLESSINGER, J. (1984).Proliferation, enhanced expression and possiblerearrangement of EGF-R in primary human braintumors of glial origin. Nature, Lond. 313, 144-146.

LIN, C. R., CHEN, W. S., KRUIGER, W., STOLARSKY, L.S., WEBER, W., EVANS, R. M., VERMA, I. M., GILL, G.N. & ROSENFELD, M. G. (1984). Expression cloning ofhuman EGF receptor complementary DNA: geneamplification and three related messenger RNAproducts in A431 cells. Science 224, 843-848.

Liu, H. T., BASERGA, R. & MERCER, W. E. (1985).Adenovirus type 2 activates cell cycle-dependent genesthat are a subset of those activated by serum. Mol.Cell. Biol. 5, 2936-2942.

LYNCH, S. A., BRUGGE, J. S. & LEVINE, J. M. (1986).Induction of altered c-src product during neuraldifferentiation of embryonal carcinoma cells. Science234, 873-876.

MADHAVAN, K., BILODEAU-WENTWORTH, D. &WADSWORTH, S. C. (1985). Family of developmental^regulated, maternally expressed Drosophila RNAspecies detected by a \-myc probe. Mol. Cell. Biol. 5,7-16.

MANESS, P. F. (1986). p p ^ ^ encoded by theprotooncogene c-src is a product of sensory neurons. J.Neurosci. Res. 16, 127-139.

MANESS, P. F., SORGE, L. K. & FULTS, D. W. (1986). Anearly developmental phase of pp60COT: expression in theneural ectoderm. Devi Biol. 117, 83-89.

MARQUARDT, H., HUNKAPILLAR, M. W., HOOD, L. E. &TODARO, G. J. (1984). Rat transforming factor type I:structure and relation to epidermal growth factor.Science 223, 1079-1081.

MASON, I., MURPHY, D. & HOGAN, B. L. M. (1985).Expression of c-fos in parietal endoderm, amnion anddifferentiating F9 teratocarcinoma cells. Differentiation30, 76-81.

MASSAGU£, J. (1985). The transforming growth factors.Trends Biochem Sci. 10, 237-241.

MATRISIAN, L. M., PATHAK, M. & MAGUN, B. E. (1982).Identification of an epidermal growth factor-relatedtransforming growth factor from rat fetuses. Biochem.Biophys. Res. Comm. 107, 761-769.

MELLON, P., PAWSON, A., BISTER, K., MARTIN, G. S. &DUESBERG, P. H. (1978). Specific RNA sequences andgene products of MC29 avian acute leukemia virus.Proc. natn. Acad. Sci. U.S.A. 75, 5874-5878.

MILBRANDT, J. (1986). Nerve growth factor rapidlyinduced c-fos mRNA in PC12 rat pheochromocytomacells. Proc. natn. Acad. Sci. U.S.A. 83, 4789-4793.

MILLER, A. D., CURRAN, T. & VERMA, I. M. (1984). c-fosprotein can induce cellular transformation: a novelmechanism of activation of a cellular oncogene. Cell 36,51-60.

MITCHELL, R. L., HENNING-CHUBB, C , HUBERMAN, E. &VERMA, I. M. (1986). c-Fos expression is neithersufficient nor obligatory for differentiation ofmonomyelocytes to macrophages. Cell 45, 497-504.

MITCHELL, R. L., ZOKAS, L., SCHREIBER, R. D. & VERMA,I. M. (1985). Rapid induction of the expression ofproto-oncogene fos during human monocyticdifferentiation. Cell 20, 209-217.

MOORE, J. P., TODD, J. A., HESKETH, T. R. & METCALF,J. C. (1986). c-fos and c-myc gene activation, ionicsignals and DNA synthesis in thymocytes. J. biol.Chem. 261, 8158-8162.

MORGAN, J. I. & CURRAN, T. (1986). Role of ion flux inthe control of c-fos expression. Nature, Lond. 322,552-555.

MOZER, B., MARLOR, R., PARKHURST, S. & CORCES, V.(1985). Characterization and developmental expressionof a Drosophila ras oncogene. Mol. Cell. Biol. 5,885-889.

MULCAHY, L. S., SMITH, M. R. & STACEY, D. W. (1985).Requirement for ras protooncogene function duringserum-stimulated growth of NIH 3T3 cells. Nature,Lond. 313, 241-243.

MULLER, R. (1983). Differential expression of cellularoncogenes during murine development and interatocarcinoma cell lines. Cold Spring Harb. Conf. CellProlif. 10, 451-468.

MOLLER, R. (1986). Proto-oncogenes and differentiation.Trends Biochem. Sci. 11, 129-132.

MOLLER, R., BRAVO, R., BURCKHARDT, J. & CURRAN, T.(1984). Induction of c-fos gene and protein by growthfactors precedes activation of c-myc. Nature, Lond. 312,716-720.

Page 20: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

468 E. D. Adamson

MOLLER, R., CURRAN, T., MOLLER, D. & GUILBERT, L.(1985). Induction of c-fos during myelomonocyticdifferentiation and macrophage proliferation. Nature,Lond. 314, 546-548.

MOLLER, R., MOLLER, D. & GUILBERT, L. (1984).Differential expression of c-fos in hematopoietic cells:correlation with differentiation of monomyelocytic cellsin vitro. EMBO J. 3, 1887-1890.

MOLLER, R., MULLER, D., VERRIER, B., BRAVO, R. &HERBST, H. (1986). Evidence that expression of c-fosprotein in amnion cells is regulated by external signals.EMBOJ. 5, 311-316.

MOLLER, R., SLAMON, J., ADAMSON, E. D., TREMBLAY, J.M., MOLLER, D., CLINE, M. J. & VERMA, I. M.(1983a). Transcription of cellular oncogenes c-ras1^ andc-fms during mouse development. Mol. Cell. Biol. 3,1062-1069.

MCLLER, R., SLAMON, D. J., TREMBLAY, J. M., CLINE, M.J. & VERMA, I. M. (1982). Differential expression ofcellular oncogenes during pre- and post-nataldevelopment of the mouse. Nature, Lond. 299,640-644.

MOLLER, R., TREMBLAY, J. M., ADAMSON, E. D. &VERMA, I. M. (1983b). Tissue and cell type-specificexpression of two human c-onc genes. Nature, Lond.304, 454-456.

MOLLER, R. & VERMA, I. M. (1984). Expression ofcellular oncogenes. Curr. Top. Micr. 112, 73-115.

MOLLER, R., VERMA, I. M. & ADAMSON, E. D. (1983C).Expression of c-onc genes: c-fos transcripts accumulateto high levels during development of mouse placenta,yolk sac and amnion. EMBO J. 2, 679-684.

MOLLER, R. & WAGNER, E. (1984). Differentiation of F9'teratocarcinoma stem cells after transfer of c-fos proto-oncogene. Nature, Lond. 311, 438-442.

MUMMERY, C. L., FEUEN, A., VAN DER SAAG, P. T., VANDEN BRINK, C. E. & DE LAAT, S. W. (1985). Clonalvariants of differentiated P19 embryonal carcinomacells exhibit epidermal growth factor receptor kinaseactivity. Devi Biol. 109, 402-410.

NAU, M. M., BROOKS, B. J., BATTEY, J., SAUVILLE, E.,GAXDAR, A. F., KIRSCH, I. R., MCBRIDE, B. W.,BERTNESS, V., HOLLIS, G. F. & MINNA, J. D. (1985).L-myc; a new myc related gene amplified and expressedin human small cell lung cancer. Nature, Lond. 318,69-73.

NECKAMEYER, W. S., SHIBUYA, M., HSU, M.-T. & WANG,L.-H. (1986). Proto-oncogene c-ros codes for amolecule with structural features common to those ofgrowth factor receptors and displays tissue-specific anddevelopmentally regulated expression. Mol. Cell. Biol.6, 1478-1486.

NEER, E. J. & LOK, J. M. (1985). Partial purification andcharacterization of a pp6Cf'src related tyrosine kinasefrom bovine brain. Proc. natn. Acad. Sci. U.S.A. 82,6025-6029.

NEX0, E., HOLLENBERG, M. D., FIGUEROA, A. & PRATT,R. M. (1980). Detection of epidermal growth factor-urogastrone and its receptor during mouse fetaldevelopment. Proc. natn. Acad. Sci. U.S.A. 77,2782-2785.

NODA, M., Ko, M., OGURA, M., LIU, D. G., AMARO, T.,TAKANO, T. & IKAWA, Y. (1985). Sarcoma virusescarrying ras oncogenes induce differentiation-associatedproperties in a neuronal cell line. Nature, Lond. 318,73-75.

O'LOUGHLIN, E. V., CHUNG, M., HOLLENBERG, M.,HAYDEN, J., ZAHAVI, I. & GALL, D. G. (1985). Effectof epidermal growth factor on ontogeny of thegastrointestinal tract. Am. J. Physiol. 249, 674-678.

OPPENHEIMER, J. H. (1979). Thyroid hormone action atthe cellular level. Science 203, 971-979.

PAPKOFF, J., NIGG, E. A. & HUNTER, T. (1983). Thetransforming protein of Moloney murine sarcoma virusis a soluble cytoplasmic protein. Cell 33, 161-172.

PAPKOFF, J., VERMA, I. M. & HUNTER, T. (1982).Detection of a transforming gene product in cellstransformed by Moloney murine sarcoma virus. Cell 29,417-426.

PAWSON, T., AMID, T., HINZE, E., AUERSPERG, N.,NEAVE, N., SOBOLEWSKI, A. & WEEKS, G. (1985).Regulation of a ras-related protein during developmentof Dictyostelium discoideum. Mol. Cell. Biol. 5, 33-39.

PFEIFER-OHLSSON, S., GOUSTIN, A. S., RYDNERT, J.,WAHLSTROM, T., BJERSING, L., STEHELIN, D. &OHLSSON, R. (1984). Spatial and temporal pattern ofcellular myc oncogene expression in developing humanplacenta: Implications for embryonic cell proliferation.Cell 38, 585-596.

PFEIFER-OHLSSON, S., RYDNERT, J., GOUSTIN, A. S.,LARSSON, E., BETSHOLTZ, C. & OHLSSON, R. (1985).Cell-type specific pattern of myc proto-oncogeneexpression in developing human embryos. Proc. natn.Acad. Sci. U.S.A. 82, 5050-5054.

POPLIKER, M., SHATZ, A., AVIVI, A., ULLRICH, A.,SCHLESSINGER, J. & WEBB, C. G. (1986). Onset ofendogenous synthesis of epidermal growth factor inneonatal mice. Devi Biol. 118 (in press).

PROPER, J. A., BJORNSON, C. L. & MOSES, H. L. (1982).Mouse embryos contain polypeptide growth factor(s)capable of inducing a reversible neoplastic phenotypein nontransformed cells in culture. /. cell. Physiol. 110,169-174.

PROPST, F. & VANDE WOUDE, G. F. (1985). Expression ofc-mos proto-oncogene transcripts in mouse tissues.Nature, Lond. 315, 516-518.

PURCHIO, A. F., ERIKSON, E., BRUGGE, J. S. & ERIKSON,R. L. (1978). Specific RNA sequences and geneproducts of MC29 avian acute leukemia virus. Proc.natn. Acad. Sci. U.S.A. 75, 1567-1571.

RABBITTS, P. H., WATSON, J. V., LAMOND, A., FURSTER,A., STINSON, M. A., EVAN, G., FISCHER, W.,ATHERTON, E., SHEPPARD, R. & RABBITS, T. H. (1985).Metabolism of c-myc gene products: c-myc mRNA andprotein expression in the cell cycle. EMBO J. 4,2009-2015.

RALL, L. B., SCOTT, J., BELL, G. I., CRAWFORD, R. J.,PENSCHOW, J. D., NIALL, H. D. & COGHLAN, J. P.(1985). Mouse prepro-epidermal growth factorsynthesis by the kidney and other tissues. Nature, Lond.313, 228-231.

Page 21: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 469

RAO, C. D., IGARASHI, H., CHIU, I. M., ROBBINS, K. C.

& AARONSON, S. A. (1986). Structure and sequence ofthe human c-sis/PDGF-2 transcriptional unit. Proc.natn. Acad. Sci. U.S.A. 83, 2392-2396.

RAZIN, A., WEBB, C , SZYF, M., YISRAELI, J.,

ROSENTHAL, A., NAVEH-MANY, T., SCIAKY-GALULI, N.

& CEDAR, H. (1984). Variations in DNA methylationduring mouse cell differentiation in vivo and in vitro.Proc. natn. Acad. Sci. U.S.A. 81, 2275-2279.

REDDY, E. P., SMITH, M. J. & SRINIVASAN, A. (1983).

Nucleotide sequence of Abelson murine leukemia virusgenome: structural similarity of its transforming gene toother one gene products with tyrosine-specific kinaseactivity. Proc. natn. Acad. Sci. U.S.A. 80, 3623-3627.

REED, J. C , ALPERS, J. D., NOWELL, P. C. & HOOVER,

R. G. (1986). Sequential expression of protooncogenesduring lectin-stimulated mitogenesis of normal humanlymphocytes. Proc. natn. Acad. Sci. U.S.A. 83,3982-3986.

REES, A. R., ADAMSON, E. D. & GRAHAM, C. F. (1979).Epidermal growth factor receptors increase during thedifferentiation of embryonal carcinoma cells. Nature,Lond. 281, 309-311.

RENZ, M., NEUBERG, M., KURZ, C , BRAVO, R. &

MOLLER, R. (1985). Regulation of c-fos transcription inmouse fibroblasts: identification of DNasel-hypersensitive sites and regulatory upstream sequences.EMBOJ. 4,3711-3716.

RENZ, M., VERRIER, B., KURZ, C. & MOLLER, R. (1987).Chromatin association and DNA binding properties ofthe c-fos proto-oncogene. Nucl. Acids. Res. 15,277-292.

RESH, M. D. & ERIKSON, R. L. (1985). Highly specificantibody to Rous sarcoma virus sre gene productrecognizes a novel population of pp60VJTC and pptiff'"*molecules. / . Cell Biol. 100, 409-417.

RETTENMIER, C. W., CHEN, J. H., ROUSSEL, M. F. &

SHERR, C. J. (1985). The product of the c-frns proto-oncogene: A glycoprotein with associated tyrosinekinase activity. Science 228, 320-322.

RETTENMIER, C. W., ROUSSEL, M. F., QUINN, C. O.,

KJTCHINGMAN, G. R., LOOK, A. T. & SHERR, C. J.

(1985). Transmembrane orientation of glycoproteinsencoded by the v-fms oncogene. Cell 40, 971-981.

RIZZINO, A. (1982). Embryonal carcinoma cells releasefactors with transforming growth factor (TGF) activity./ . Cell Biol. 95, 181a.

RIZZINO, A. & BOWEN-POPE, D. F. (1985). Production ofPDGF-like factors by embryonal carcinoma cells andbinding of PDGF to their endoderm-like differentiatedcells. Devi Biol. 110, 15-22.

ROBBINS, K. C , ANTONIADES, H. N., DEVARE, S. G.,

HUNKAPILLER, M. W. & AARONSON, S. A. (1983).Structural and immunological similarities betweensimian sarcoma virus gene product(s) and humanplatelet-derived growth factor. Nature, Lond. 305,605-609.

ROBERTS, A. B., ANZANO, M. A., LAMB, L. C , SMITH, J.

M. & SPORN, M. B. (1981). New class of transforminggrowth factors potentiated by epidermal growth factor:

Isolation from non-neoplastic tissues. Proc. natn. Acad.Sci. U.S.A. 78, 5339-5343.

ROBERTS, A. B. & SPORN, M. B. (1985). Transforminggrowth factors. Cancer Surveys 4, 683-705.

ROSSANT, J., SANFORD, J. P., CHAPMAN, V. M. &ANDREWS, G. K. (1986). Undermethylation ofstructural gene sequences in extraembryonic lineages ofthe mouse. Devi Biol. 117, 567-573.

RUPPERT, C , GOLDWTTZ, D. & WILLE, W. (1986). Proto-oncogene myc is expressed in cerebellar neurons atdifferent developmental stages. EMBOJ. 5, 1897-1901.

ROTHEK, V., WAGNER, E. F. & MOLLER, R. (1985).Analysis of the differentiation-promoting potential ofinducible c-fos genes introduced into embryonalcarcinoma cells. EMBOJ. 4,1775-1781.

SADLER, S. E., SCHECHTER, A. L., TABIN, C. J. &MOLLER, J. L. (1986). Antibodies to the ras geneproduct inhibit adenylate cyclase and accelerateprogesterone-induced cell division in Xenopus laevisoocytes. Mol. Cell. Biol. 6, 719-722.

SALIDO, E. C , BARAJAS, L., LECHAGO, J., LABORDE, N. P.& FISHER, D. A. (1986). Immunocytochemicallocalization of epidermal growth factor in mousekidney. J. Histochem. Cytochem. 34, 1155-1160.

SAMBUCETTI, L. C. & CURRAN, T. (1986). The/os proteincomplex is associated with DNA in isolated nuclei andbinds to DNA cellulase. Science 234, 1417-1419.

SCHECHTER, A. L., HUNG, M. C, VAIDYANATHAN, L.,WEINBERG, R. A. & YANG-FEND, T. L., FRANCKE, U.,ULLRICH, A. & COUSSENS, L. (1985) The neu gene: Anerb-B-homologous gene distinct from and unlinked tothe gene encoding the EGF receptor. Science 229,976-978.

SCHECHTER, A. L., STERN, D. F., VAIDYANATHAN, L.,DECKER, S. J., DREBIN, J. A., GREEN, M. I. &WEINBERG, R. A. (1984). The neu. oncogene: an erb-B-related gene encoding a 185,000Mr tumour antigen.Nature, Lond. 312, 513-516.

SCOTT, J., URDEA, M., QUIROGA, M., SANCHEZ-PESCADOR,R., FONG, N., SELBY, M., RUTTER, W. J. & BELL, G. I.(1983). Structure of a mouse submaxillary mRNAencoding epidermal growth factor and seven relatedproteins. Science 111, 236-239.

SEGAL, D., SHILO, B.-Z. (1986). Tissue localization ofDrosophila melanogaster ras transcript duringdevelopment. Mol. Cell. Biol. 6, 2241-2248.

SEIFERT, R. A., SCHWARTZ, S. M. & BOWEN-POPE, D. F.(1984). Developmentally regulated production ofplatelet-derived growth factor-like molecules. Nature,Lond. 311, 669-671.

SEJERSEN, T., BJORKLUND, H., SOMEGI, J. & RINGERTZ,N. R. (1986). N-myc and c-src genes are differentiallyregulated in PCC7 embryonal carcinoma cellsundergoing neuronal differentiation. J. cell. Physiol.127, 274-280.

SEJERSEN, T., SUMEGI, J. & RINGERTZ, N. R. (1985).Expression of cellular oncogenes in teratocarcinoma-derived cell lines. Expl Cell Res. 160, 19-30.

SHEINESS, B. & GARDINIER, M. (1984). Expression ofproto-oncogene (proto-myb) in hemopoietic tissues ofmice. Mol. Cell. Biol. 4, 1206-1212.

Page 22: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

470 E. D. Adamson

SHERR, C. J., RETTENMEIER, G. W., SALA, R., ROUSSEL,M. F., LOOK, A. T. & STANLEY, E. R. (1985). The c-fms proto-oncogene product is related to the receptorfor the mononuclear phagocyte growth factor, CSF-1.Cell 40, 665-675.

SIMMEN, F. A., GOPE, M. L., SCHULZ, T. Z., WRIGHT, D.A., CARPENTER, G. & O'MALLEY, B. W. (1984).Isolation of an evolutionarily conserved epidermalgrowth factor receptor cDNA from human A431 cells.Biochem. Biophys. Res. Comm. 124, 125-132.

SIMON, M. A., DREES, B., KORNBERG, T. & BISHOP, J. M.(1985). The nucleotide sequence and the tissue-specificexpression of Drosophila c-src. Cell 42, 831-840.

SINKOVICS, J. G. (1984). Retroviral and human cellularoncogenes. Ann. Clin. lab. Sci. 14, 343-354.

SLAMON, D. J. & CLINE, M. J. (1984). Expression ofcellular oncogenes during embryonic and fetaldevelopment of the mouse. Proc. natn. Acad. Sci.U.S.A. 81, 7141-7145.

SMELAND, E., GODAL, T., RUUD, E., BEISKE, K.,FUNDERRUD, S., CLARK, E. A., PFEIFER-OHLSSON, S. &OHLSSON, R. (1985). The specific induction of mycproto-oncogene expression in normal human B cells isnot a sufficient event for acquisition of competence toproliferate. Proc. natn. Acad. Sci. U.S.A. 82,6255-6259.

SODERQUIST, A. M. & CARPENTER, G. (1986). Biosynthesisand metabolic degradation of receptors for epidermalgrowth factor. / . Membrane Biol. 90, 97-105.

SORGE, L. K., LEVY, B. T. & MANESS, P. F. (1984).W&Fsrc is developmentally regulated in the neuralretina. Cell 36, 249-256.

STEWART, T. A., BELLVE, A. R. & LEDER, P. (1984).Transcription and promoter usage of the myc gene innormal somatic and spermatogenic cells. Science 226,707-710.

STROMBERG, K., PIGOTT, D. A., RANCHALIS, J. E. &TWARDZIK, D. R. (1982). Human term placentacontains transforming growth factors. Biochem.Biophys. Res. Comm. 106, 354-361.

STROMBERG, K. & TWARDZIK, D. R. (1985). A j3-typetransforming growth factor, present in conditioned cellculture medium independent of cell transformation,may derive from serum. J. Cell Biochem. 27, 443-448.

STUDZINSH, S., BRELVI, Z. S., FELDMAN, S. C. & WATT,R. A. (1986). Participation of c-myc protein in DNAsynthesis of human cells. Science 234, 467—470.

SUGIMOTO, Y., WHITMAN, M., CANTLEY, L. C. & ERIKSON,R. L. (1984). Evidence that the Rous sarcoma virustransforming gene product phosphorylatesphosphatidylinositol and diacylglycerol. Proc. natn.Acad. Sci. U.S.A. 81, 2117-2121.

SWEET, R. W., YOKOYAMA, S., KAMATA, T., FERAMISCO, J.R., ROSENBERG, M. & GROSS, M. (1984). The productof ras is a GTPase and the T24 oncogenic mutant isdeficient in this activity. Nature, Lond. 311, 273-275.

TAINSKY, M. A., COOPER, C. S., GIOVANELLA, B. C. &VANDE WOUDE, G. F. (1984). An activated rasN gene:detected in late but not early passage human PA1teratocarcinoma cells. Science 225, 643—645.

TAKEYA, T. & HANAFUSA, H. (1983). Structure andsequence of the cellular gene homologous to the RSVsrc gene and the mechanism for generating thetransforming virus. Cell 32, 881-890.

TAM, J. P., MARQUARDT, H., ROSBERGER, D. F., WONG,T. W. & TODARO, G. J. (1984). Synthesis ofbiologically active rat transforming growth factor a.Nature, Lond. 309, 376-378.

TEMELES, G. L., GIBBS, J. B., D'ALONZO, J. S., SIGAL, I.S. & SCOLNICK, E. M. (1985). Yeast and mammalian rasprotein have conserved biochemical properties. Nature,Lond. 313, 700-703.

THOMPSON, C. B., CHAKONER, P. B., NEIMAN, P. E. &GROUDINE, M. (1985). Levels of c-myc oncogenemRNA are invariant throughout the cell cycle. Nature,Lond. 314, 363-366.

THOMPSON, D. M. & GILL, G. N. (1985). The EGFreceptor: structure, regulation and potential role inmalignancy. Cancer Surveys 4, 767-788.

TODOKORO, K. & IKAWA, Y. (1986). Sequential expressionof proto-oncogenes during a mouse erythroleukemiacell differentiation. Biochem. Biophys. Res. Comm. 135,1112-1118.

TRAMONTANO, D., CHIN, W. W., MOSES, A. C. & INGBAR,S. H. (1986). Thyrotropin and dibutyryl cyclic AMPincrease levels of c-myc and c-fos mRNAs in culturedrat thyroid cells. / . biol. Chem. 261, 3919-3922.

TREISMAN, R. (1985). Transient accumulation of c-fosRNA following serum stimulation requires a conserved5' element and c-fos 3' sequences. Cell 42, 889-902.

TSUCMDA, N., RYDER, T. & OHTSUBO, E. (1982).Nucleotide sequence of the oncogene encoding the p21transforming protein of Kirsten murine sarcoma virus.Science 217, 937-939.

TURLEY, E. A., HOLLENBERG, M. D. & PRATT, R. M.(1985). Effect of epidermal growth factor/urogastroneon glycosaminoglycan synthesis and accumulation invivo in the developing mouse palate. Differentiation 28,279-285.

TWARDZIK, D. R. (1985). Differential expression oftransforming growth factor ex during prenataldevelopment of the mouse. Cancer Res. 45, 5413-5416.

TYLER, M. S. & PRATT, R. M. (1980). Effect of EGF onsecondary palatal epithelium in vitro: tissue isolationand recombination studies. / . Embryol. exp. Morph. 58,93-106.

ULLRICH, A., COUSSENS, L., HAYFLICK, J. S., DULL, T. J.,GREY, A., TAM, A. W., LEE, J., YARDEN, Y.,LlBERMAN, T. A . , SCHLESSINGER, J. , DOWNWARD, J. ,MAYES, E. L. V., WHITTLE, N., WATERFIELD, M. D. &SEEBURG, P. H. (1984). Human epidermal growthfactor receptor cDNA sequence and aberrantexpression of the amplified gene in A431 epidermoidcarcinoma cells. Nature, Lond. 309, 418-425.

USHIRO, H. & COHEN, S. (1985). Identification ofphosphotyrosine as product of EGF activated proteinkinase in A431 cell membranes. / . biol. Chem. 255,8363-8365.

VAN BEVEREN, C , GALLESHAW, J. A., JONAS, V., BERNS,A. J. M., DOOLITTLE, R. F., DONOGUE, D. J. & VERMA,I. M. (1981). Nucleotide sequence and formation of the

Page 23: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no

Oncogenes in development 471

transforming gene of a mouse sarcoma virus. Nature,Lond. 289, 258-262.

VAN BEVEREN, C , VAN STRAATEN, F., CURRAN, T.,

MULLER, R. & VERMA, I. M. (1983). Analysis of FBJ-MuSV proteins and c-fos (mouse) gene reveals thatviral and cellular fos gene products have differentcarboxy termini. Cell 32, 1241-1255.

VARMUS, H. E. (1984). The molecular genetics of cellularoncogenes. A. Rev. Genet. 18, 553-612.

VERRIER, B., MULLER, D., BRAVO, R. & MULLER, R.

(1986). Wounding a fibroblast monolayer results in therapid induction of the c-fos proto-oncogene. EM BO J.5, 913-917.

VILETTE, D., EMANOIL-RAVIER, R., TOBALY, J. & PERIES,

J. (1985). Studies on four cellular proto-oncagenes andtheir expression in PCC4 embryonal carcinoma cells:Amplification of c-Ki-ros oncogene. Biochem. Biophys.Res. Comm. 128, 513-519.

WATERFIELD, M. D., SCRACE, G. T., WHITTLE, N.,

STROOBANT, P., JOHNSSON, A., WASTESON, A.,

WESTERMARK, B., HELDIN, C.-H., HUANG, J. S. &DEUEL, T. F. (1983). Platelet-derived growth factor isstructurally related to the putative transforming proteinp28m of simian sarcoma virus. Nature, Lond. 304,35-39.

WATT, R. A., SHATZMAN, A. R. & ROSENBERG, M.

(1985). Expression and characterization of the humanc-myc DNA-binding protein. Mol. Cell. Biol. 5,448-456.

WEINBERG, R. A. (1984). Cellular oncogenes. TrendsBiochem. Sci. 9, 131-133.

WEINBERGER, C , HOLLENBERG, S. M., ROSENFELD, M. G.

& EVANS, R. M. (1985). Domain structure of humanglucocorticoid receptor and its relationship to the v-erbA oncogene product. Nature, Lond. 318, 670-672.

WEINBERGER, C , THOMPSON, C. C , ONG, E. S., LEBO,

R., GRUOL, D. J. & EVANS, R. M. (1986). The c-Erb-Agene encodes a thyroid hormone receptor. Nature,Lond. 324, 641-646.

WELLER, A., MEEK, J. & ADAMSON, E. D. (1987).Preparation and properties of monoclonal andpolyclonal antibodies to mouse epidermal growth factor(EGF) receptors: Evidence for cryptic EGF receptorsin embryonal carcinoma cells. Development(submitted).

WITTE, O. N., DASGUPTA, A. & BALTIMORE, D. (1980).Abelson murine leukemia virus protein isphosphorylated in vitro to form phosphotyrosine.Nature, Lond. 283, 826-828.

WITTE, O. N., ROSENBERG, N. & BALTIMORE, D. (1979).Preparation of syngeneic tumor regressor serumreactive with the unique determinants of the Abelsonmurine leukemia virus-encoded P120 protein at the cellsurface. / . Virol. 31, 776-784.

WITTE, O. N., ROSENBERG, N., PASKIND, M., SHIELDS, A.& BALTIMORE, D. (1978). Identification of an Abelsonmurine leukemia virus encoded protein present intransformed fibroblasts and lymphoid cells. Proc. natn.Acad. Sci. U.S.A. 75, 2488-2492.

WYKE, J. (1983). From c-src to v-src. Nature, Lond. 304,491-492.

Xu, Y.-H., RICHERT, N., ITO, S., MERLINO, G. T. &PASTAN, I. (1984). Characterization of epidermalgrowth factor receptor gene expressed in malignant andnormal human cell lines. Proc. natn. Acad. Sci. U.S.A.81, 7708-7712.

YOUNG, P. R. & TILGHMAN, S. M. (1984). The inductionof a-fetoprotein synthesis in differentiating F9teratocarcinoma cells is accompanied by a genome-wideloss of DNA methylation. Mol. Cell. Biol. 4, 898-907.

ZIMMERMAN, K. A., YANCOPOULOS, G. D., COLLUM, R.G., SMITH, R. K., KOHL, N. E., DENIS, K. A., NAU,M. M., WITTE, O. N., TORAN-ALLERAND, D., GEE, C.E., MINNA, J. D. & ALK, F. W. (1986). Differentialexpression of myc family genes during murinedevelopment. Nature, Lond. 319, 780-783.

ZULLO, J. N., COCHRAN, B. H., HUANG, A. S. & STILES,C. D. (1985). Platelet-derived growth factor anddouble-stranded ribonucleic acids stimulate expressionof the same genes in 3T3 cells. Cell 43, 793-800.

Page 24: Oncogenes in developmentOncogenes in development EILEEN D. ADAMSON La Jolla Cancer Research Foundation, 10901 North Torrey Road, La Jolla, Pines CA 92037, USA Synopsis Section no