Pathology of Chickens Infected with Avian Nephroblastoma Virus MAV-2(N)

Page created by Deborah Hunter
 
CONTINUE READING
INFECTION   AND IMMUNITY, Feb. 1980,   p. 501-512                                                       Vol. 27, No. 2
0019-9567/80/02-0501/12$02.00/0

  Pathology of Chickens Infected with Avian Nephroblastoma
                      Virus MAV-2(N)
                            SUSAN L. WATTSt AND RALPH E. SMITH*
 Department of Microbiology and Immunology, Duke University Medical Center, Durham, North Carolina
                                                          27710

               A nephroblastoma-inducing myeloblastosis-associated virus, MAV-2(N), de-

                                                                                                                         Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
            rived from avian myeloblastosis virus was characterized with respect to biochem-
            ical composition and avian pathogenesis. Purified fibroblast-grown virus con-
            tained the same size 35S ribonucleic acid and the same relative amounts of viral
            polypeptides as another myeloblastosis-associated virus inducing predominantly
            osteopetrosis MAV-2(0). Plaque-purified MAV-2(N) induced a 76 to 93% inci-
            dence of nephroblastoma and a 3 to 50% incidence of osteopetrosis in SPAFAS
            and line 15 x 7 chickens: the oncogenic spectrum and the onset of nephroblastoma
            varied with the line of chicken and the route of injection. Renal neoplasms were
            manifest in chickens older than 2 months and grew to a massive size. Furthermore,
            29% of control chickens housed with MAV-2(N)-infected chickens demonstrated
            nephroblastoma. MAV-2(N)-infected chickens had growth rates and blood packed
            cell volumes comparable to those of uninfected chickens. Infected chickens 2
            months of age had increased kidney, liver, and spleen weights; tumor-bearing
            chickens 3 to 4 months of age had increased liver, lung, brain, pancreas, and bone
            weights. The concentration of albumin was decreased and the concentration of
            gamma globulin was increased in the serum of MAV-2(N)-infected chickens.
            Analysis of the sera of nephroblastoma-bearing chickens for virus and antibody
            showed that three states existed: (i) high levels of neutralizing antibody, (ii) high
            levels of virus, and (iii) simultaneous presence of both at low levels. The patho-
            logical and virological features of MAV-2(N) which distinguish it from MAV-2(0)
            are discussed.

  The standard strain of avian myeloblastosis                    cells, bone marrow cells, and macrophages in
virus (AMV), BAI strain A, primarily induces                     vitro, they cannot replicate in chicken embryo
myeloblastosis, but also induces osteopetrosis,                  fibroblast (CEF) cultures (6, 31). However,
nephroblastoma, lymphoid leukosis, and other                     nephroblastoma and osteopetrosis viruses can
neoplasms in infected chickens (4). Efforts to                   replicate in CEF and cause plaques characteris-
select for the induction of a single type of neo-                tic of subgroup B leukosis viruses (17, 35, 45).
plasm led to the isolation of several viral species              Yet morphological transformation of any cell
from standard AMV: a subgroup A myeloblas-                       type in vitro has not been demonstrated by the
tosis-associated virus MAV-1 inducing a high                     latter type of viruses. In vivo standard AMV
incidence of osteopetrosis (45); a subgroup B                    induces myeloblastosis within 2 weeks after in-
leukosis virus inducing predominantly osteope-                   fection (4), whereas nephroblastoma, osteope-
trosis MAV-2(0) (45); a subgroup B leukosis                      trosis, or lymphoid leukosis become manifest in
virus inducing primarily nephroblastoma (25,35,                  chickens infected with standard AMV or with
50); and leukemogenic viruses of subgroups A                     myeloblastosis-associated viruses weeks or
and B, respectively, AMV-A and AMV-B (22,                        months after infection (10, 35, 36, 45).
32).                                                                MAV strains primarily inducing osteopetrosis
   Evidence suggests that the nephroblastoma-                    were isolated by endpoint dilution of superna-
and osteopetirosis-inducing leukosis viruses are                 tant fluids obtained from cell cultures of stan-
nondefective helper viruses providing glycopro-                  dard AMV-induced chicken tumors of standard
tein envelope components for the replication-                    AMV-treated fibroblasts (45). Subgroup B MAV
defective leukemogenic viruses in standard                       strains inducing a high incidence of nephroblas-
AMV (30). Although the leukemogenic compo-                       toma were isolated by two routes: (i) passage of
nents have been shown to transform yolk sac                      standard AMV-induced nephroblastoma ex-
                                                                 tracts in vivo (50) and (ii) transfection ofhamster
   t Present address: Department of Microbiology, University     fibroblasts (25) or CEF (35) in vitro with stan-
of Minnesota, Minneapolis, MN 55455.                              dard AMV-induced leukemic myeloblast deoxy-
                                                               501
502      WATTS AND SMITH                                                                           INFECT. IMMUN.
 ribonucleic acid (DNA). The virus strain isolated           facility. Infected and uninfected birds in the same
 by Ogura et al. (35, 36) has been cloned in our            room were      kept in separate cages. Birds were fed
 laboratory by plaque purification: the present              Purina Growena Chow and water ad libitum. For
 investigation reports the biochemical and path-             experimental purposes, or when incapacitated by tu-
                                                             mor or other disease, chickens were sacrificed by
 ogenetic characterization of this cloned virus              exsanguination. All chickens were subjected to post-
 MAV-2(N).                                                   mortem examination.
   Avian nephroblastoma is a renal tumor of                     Chicken body weight and organ weight deter-
both epithelial and mesenchymal cell types man-              minations. Birds were weighed on a Harvard labo-
ifesting distorted morphology and abnormal dif-              ratory balance. Organs excised from birds sacrificed by
ferentiation. The avian tumor morphology is                  exsanguination were weighed on a Mettler analytical
comparable to that of Wilms' tumor of humans                balance.

                                                                                                                        Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
(33) as well as that of nephroblastomas found in               Extraction of virus from infected chicken or-
rabbits, swine, cattle, sheep, dogs, and cats (7,           gans. Extracts from organs excised from infected
14, 24, 38). Histopathological studies of avian             chickens were prepared as follows: Organs were
                                                            washed with Tris buffer or phosphate-buffered saline
nephroblastoma note the presence of all com-                (0.15 M NaCl, 2.7 mM KCl, 1.5 mM KH2PO4, 8 mM
ponents of normal kidney in various states of               Na2HPO4, 0.9 mM CaCl2, 0.5 mM MgCl2.6H20), then
organization and differentiation as well as occa-           minced with a scalpel and spatula or homogenized
sional evidence of cartilage, bone, keratinized             with a Virtis "23" homogenizer in 0.1 to 0.5 ml of Tris
epithelium, and sarcoma. Thus, an embryonic                 buffer per g. Cells were sedimented by centrifugation
rest residual in the adult kidney has been sug-             at 800 x g for 15 min at 40C, and the supernatant fluid
gested as the origin of the nephroblastoma (5,              was frozen at -70°C until used for virus titration by
21). However, the target cell and mechanism of              plaque assay.
the viral-induced transformation are unknown.                 Blood packed cell volume (PCV). Blood samples
The present study was undertaken to elucidate               were    collected in heparinized 50-,l microhematocrit
                                                             tubes and centrifuged in a Clay Adams microhemato-
the pathological manifestations of MAV-2(N)                  crit centrifuge. Packed cell volume for each sample
infection by investigating biological characteris-           was calculated by comparison to a standard scale.
tics of MAV-2(N)-infected chickens.                              Serum protein electrophoresis. Serum protein
   (This investigation is part of a dissertation             electrophoretic analyses were performed for all sam-
submitted by S.L.W. to the Department of Mi-                 ples in triplicate by using a Beckman Microzone elec-
crobiology and Immunology, Duke University                   trophoresis system, model R-101 (3). Serum samples
Medical Center, for the Ph.D. degree.)                       of 0.25 pl were applied to cellulose acetate membranes
                                                             and subjected to 4.5 mA at 250 V for 20 min in sodium
           MATERIALS AND METHODS                             barbital (B-2) buffer, pH 8.6. Membranes were stained
                                                             in Ponceau red S, then cleared in dilute glacial acetic
    Eggs. Fertile chicken eggs certified free of chicken     acid and ethanol. Membrane scans were performed by
 helper factor and avian leukosis virus group-specific       using a Quick-Scan, Jr., interfaced with a Digital PDP-
 antigen were obtained from SPAFAS, Inc., Norwich,           11/10 computer. Areas under the peaks were inte-
 Conn. Fertile eggs of line 15 x 7 were obtained from        grated and expressed relative to the total area scanned.
 the Regional Poultry Research Laboratory, East Lan-         Serum protein concentration was determined by the
sing, Mich. Eggs were incubated at 37.50C in a humid-        method of Lowry et al. (27).
 ified incubator.                                               Immunization of chickens with virus. Unin-
    Chickens-infection and maintenance. SPA-                 fected normal adult chickens were injected intrave-
FAS and line 15 x 7 chickens were infected with 105          nously with 0.1 to 0.5 ml of virus in Tris buffer con-
plaque-forming units (PFU) of MAV-2(N) (0.1 ml)              taining 10% calf serum and reinjected by the same
produced in tissue culture or mock-infected with 0.1        procedure 3 to 4 weeks later. Approximately 2 months
ml of tris(hydroxymethyl)aminomethane (Tris) buffer          after the primary injection, the chickens were sacri-
 (0.14 M NaCl, 0.025 M Tris-hydrochloride, pH 7.4, 5        ficed by exsanguination, and the serum was stored at
mM glucose, 100 U of penicillin per ml, 100 ig of           -70°C for titration of serum antibody by the neutral-
streptomycin per ml, 0.7 mM Na2HPO4) containing             ization assay of Ishizaki and Vogt (23).
10% (vol/vol) calf serum by one of two routes: (i)              Statistical analysis. Values were reported as
infection of a chorioallantoic membrane vein of 12-         mean ± standard error for each determination. Levels
day-old embryos (2), or (ii) intraperitoneal infection of   of significance (P) were calculated by Student's two-
1-day-old hatched chicks. MAV-2(0) was adminis-             tailed t test. P values refer to the difference in param-
tered at 104 PFU per embryo [a lower dose than MAV-         eters between MAV-2(N)-infected and uninfected
2(N)] to the same lines of chickens, because 105 PFU        chickens of the same age.
of MAV-2(0) per embryo led to high embryo mortality            Virus sources and derivations. A nephroblas-
and early death of the hatched chick due to severe          toma-inducing virus derived by transfection of CEF in
osteopetrosis. One day before hatching, eggs were           culture with DNA from AMV-induced leukemic mye-
transferred to humidified hatchers maintained at            loblasts (35) was received from Heinz Bauer, Institut
37.50C, separate hatchers being used for infected and       fur Virologie, Justus-Liebig Universitat, Giessen, W.
control eggs. Birds infected with different viruses were    Germany. This virus, cloned three times by plaque
maintained in separate rooms of an animal isolation         purification and propagated in our laboratory in SPA-
VOL. 27, 1980                                  AVIAN NEPHROBLASTOMA VIRUS PATHOLOGY                               503

FAS CEF (C/E), will hereafter be referred to as MAV-             Polyacrylamide gel electrophoresis of viral
2(N), denoting that it is a myeloblastosis-associated         RNA. Viral 35S RNA was analyzed by electrophoresis
virus of subgroup B inducing nephroblastoma. MAV-             in 2.1% (wt/vol) acrylamide-0.6% (wt/vol) agarose-6
2(0) (45) has been well characterized in our laboratory       M urea gels (15). Gel slices were solubilized overnight
(3).                                                          at 370C in Protosol/toluene/water (9:10:1, vol/vol),
    All other viruses were obtained as follows: Prague        and radioactivity in each sample was determined in a
B strain of Rous sarcoma virus (PR.RSV-B) from R.             toluene-based liquid scintillation cocktail.
R. Friis, Institut fur Virologie, Justus-Liebig Univer-                          RESULTS
sitat, Giessen, W. Germany; Prague C strain of Rous
sarcoma virus (PR.RSV-C), and ring-necked pheasant              Biochemical composition of MAV-2(N).
virus (RPV) from P. K. Vogt, University of Southern           The polypeptide composition of purified MAV-
California, Los Angeles; and AMV from J. W. Beard,            2(N) relative to that of other avian retroviruses
Life Sciences, Inc., St. Petersburg, Fla. MAV-2(N) and        was investigated by SDS-polyacrylamide gel

                                                                                                                           Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
MAV-2(0) pseudotypes of Rous sarcoma virus (RSV)              electrophoresis. Electrophoretically separated
were produced by infection of Bryan strain RSV(-)
CEF with MAV-2(N) or MAV-2(0). Cells transformed              MAV-2(N) polypeptides comigrated with the
by RSV(-) were a generous gift of H. Hanafusa,                gp85, gp37, p27, p19, p15/plO, and p12 of MAV-
Rockefeller University, New York, N.Y.                        2(0), AMV, and PR.RSV-C. Although these
    Cell culture. Primary CEFs were prepared from             polypeptides were not further characterized,
 11-day-old SPAFAS embryos as described by Vogt               their identity was assumed on the basis of elec-
 (49) and propagated in supplemented Ham F10 me-              trophoretic mobility. A comparison of the rela-
dium (42). Secondary CEFs seeded into tissue culture          tive amounts of polypeptides in each virus was
dishes or roller bottles were infected for virus produc-      achieved by using gel scans of Coomassie bril-
tion at an input multiplicity of infection of 0.01 to 0.10    liant blue-stained protein. The ratio of gp85/p27
PFU or focus-forming units per cell.
    Plaque assays were performed by the method of             provided a further index of the relative amounts
Graf (17). The focus assay of Vogt (49) was used for          of gp85 and p27 polypeptides per virion. The
assaying transforming avian sarcoma viruses (ASVs).           relative amounts of MAV-2(N) polypeptides cor-
 Antiviral antibody was assayed by plaque or focus            responded completely with the values obtained
 neutralization as described by Ishizaki and Vogt (23).       for MAV-2(0) polypeptides. MAV-2(N), as well
 Complete neutralization was scored as 90% reduction          as MAV-2(0) (48), contained more stained ma-
 in plating efficiency.                                       terial identifiable as gp85 than did AMV or
     Large-scale virus production. Infected cells were        PR.RSV-C (Table 1).
 grown in roller bottles for production of large amounts         The size of MAV-2(N) 35S RNA was com-
 of virus for ribonucleic acid (RNA) and protein isola-       pared to that of MAV-2(0) by electrophoresis
 tion. Roller bottles containing cells infected with
 MAV-2(N), MAV-2(0), and PR.RSV-C were fed and                 on the same gel. The 70S RNA from each virus
 harvested every 2 h on a Bellco Smith-Kozoman Au-            was labeled, extracted, heat denatured, and sub-
 toharvester (41, 44).
     Virus concentration, purification, and label-                  TABLE 1. Comparison of the polypeptide
 ing. Virus was concentrated by pelleting and purified         composition of MAV-2(N) with that of other avian
 on sucrose gradients as previously described (41). Vi-                           oncovirusesa
 rus pellets were suspended in buffer and stored at                                  % of total polypeptideb
  -70°C. Viral RNA was labeled with 10 mCi per roller          Polypeptide
  bottle of [5,6-3H]uridine (40 to 50 Ci/mmol) by adding         position   MAV-2- MAV-2-           AMV PR.RSV-
  the radioisotope in culture medium to confluent roller                      (N)        (0)                  C
  bottle cultures. Supernatant fluids were harvested 24           gp85        8.68      10.04        2.60    2.26
  h after addition of radioisotope and every 12 h there-          gp37        2.24       2.43        3.37    3.21
  after. Virus was labeled with carrier-free [32P]ortho-              p27       22.43    23.37      26.08      22.11
  phosphate as previously described (46).                           p19         14.59    14.02      16.27      15.27
     Viral polypeptide polyacrylamide gel electro-                  p12         14.72    14.77      15.93      15.25
  phoresis. Viral polypeptides were analyzed by elec-            p15, plO        9.28     7.73      12.80      12.95
  trophoresis in slab gels of 10% (wt/vol) acrylamide-          gp85/p27C        0.39     0.43       0.10       0.10
  0.1% (wt/vol) sodium dodecyl sulfate (SDS) (8, 48).
  Gels stained with Coomassie brilliant blue were
                                                                 a
                                                                     SDS-polyacrylamide gel electrophoresis analyses
  scanned on a Quick-Scan, Jr., interfaced with a digital     of purified PR.RSV-C, MAV-2(N), MAV-2(0), and
  PDP-11/10 computer. The peaks of optical density            AMV were performed in triplicate. Gels were stained
  obtained in the scans were integrated by the computer       with Coomassie brilliant blue and scanned, and peak
  relative to the total stained material and the amount       areas were determined by computer integration.
                                                                 b The mean value of percent stained material mi-
   of protein applied to the gel.
      Viral RNA purification. Viral RNA was purified          grating in each polypeptide position relative to the
   by SDS-pronase treatment and phenol extraction of          total stained material on the gel is given for each virus.
                                                                 c The ratio of gp85 to p27 was calculated as an index
   virus pellets (11, 13). Viral 70S RNA was isolated by
   rate-zonal gradient centrifugation in gradients of 15 to   of the relative amounts of these polypeptides in each
   30% (wt/vol) sucrose (13).                                  virus.
504      WATTS AND SMITH                                                                                             INFECT. IMMUN.
                  AND SMITH
          504 WATTS                                                                                          IMMUN.~~~.if
                                                                                                               INFECT.
jected to electrophoresis as described in Mate-                                         _   4       o0
rials and Methods. The MAV-2(N) 35S RNA
peak comigrated precisely with that of MAV-                         >   2           c
2(0).
   Pathology of chickens infected with                              4       0   >

MAV-2(N). Chickens infected with plaque-pu-                             o               tL_         t-
rifled CEF-grown MAV-2(N) manifested an in-                         c                           o
cidence of nephroblastoma greater than 75% and
an incidence of osteopetrosis less than 50%, and                           _ o 00
none of the other neoplasms associated with                                                      o0
 AMV (myeloblastosis, lymphoid leukosis) (Ta-                      tic  C
 ble 2). The nephroblastomas induced by MAV-             S n                   W MLo

                                                                                                                                      Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
                                                                                    0

 2(N) appeared identical by gross and histological       W    f >.
 examination to those described after induction
                                                                     CC +1 +I+   -H-
 by AMV or AMV-derived virus strains (21, 26,                  z            CD 00
 36). The nephroblastomas consisted of abnor-
 mally differentiated kidney tissue containing ele-  S                        t        °            o
ments of sarcoma, carcinoma, chondroma, os-         .q .c     V.
 teoid, and keratinized tissue. These noninvasive              Z                o
 neoplasms were multifocal in origin and pro-
 gressed to form massive, thickly encapsulated 3J                        0N                                c
 structures. The tumors varied in their composi-     2!                     V M      D         - -O
 tion of fluid-filled cysts, solid tissue, necrotic          0o
 areas, and hemorrhagic areas. The largest ob- c5 _
 served tumor (369 g) exceeded one-fifth the body                -Q LO_c
 weight of the chicken. The osteopetrosis ob- ;                                            CC
 served with MAV-2(N) infection was of a later                                   0 4n
 onset and a more peripheral body distribution,     X                         C

 but otherwise was identical to that observed                                                         o                     4
 after infection with MAV-2(0) (3, 45).                    rC c14

    MAV-2(N)-infected chickens manifested sev-         -s O                     o
 eral other abnormalities, some of which could be    W                          _S o2 'C .      -

attributed to virus infection or to the presence     S o           0 CC
of nephroblastoma. Chickens dying before age
60 days died of cannibalism, anemia, other infec-
                                                        00
                                                     ;: la
                                                             _ .c
tions, intra-abdominal hemorrhage, or unknown                                       -          . .
                                                                          2 0 Mc q L
causes (Table 2). Chickens dying after age 60                     2           oo
                                                                              0
days also demonstrated a large incidence of can-                   O.                      .          oJ >, t0 3 3
nibalism and were frequently observed to man-               1'   z                  <   0
                                                                                         5-                  o
ifest intradermal petechial hemorrhages of un-                                     ^ 2c. p0              4

known significance. Cannibalism was rarely ob- 3                                                 o
served in uninfected chickens and chickens in-                                                    Ho > _o.0
fected with other viruses, indicating that feeding,         o
light, and confinement conditions did not con-              _          _0
                                                                        - o
tribute to the high rate of cannibalism seen in                                   in          o              5   4

MAV-2(N)-infected chickens. The incidence of                                  W        o -
cannibalism was recorded as a manifestation of                   8                                     O
unknown etiology associated with MAV-2(N)                                                     .2
infection. When the size of the nephroblastoma              I= U        N   ,                          0 °
caused abdominal obstruction, chickens mani-                o0          4
fested ascites, diarrhea, intra-abdominal hem-                                                    co         0  ow,
orrhage, and paralysis.                                       O=         .-
   Effect of chicken strain and route of in-
fection on the oncogenic spectrum of MAV-
                                                           o:           ->    -.+1          3
                                                                                                       cc

2(N). The relative incidence of nephroblastoma                 O          C/X           .C    C> t6iC.00 .g=0) s>       _

and osteopetrosis varied with the chicken line                .c
and route of injection used (Table 2). The inci-                                    -x
dence of nephroblastoma was approximately                                 a co
                                                                                        a
                                                                                                    o
VOL. 27, 1980                            AVIAN NEPHROBLASTOMA VIRUS PATHOLOGY                        505

80% in SPAFAS chicks and approximately 90%            relative to the total body weight of each bird
in line 15 x 7 chickens. The similarity of neph-      (Table 3). At 30 days of age, the only significant
roblastoma incidence in chickens infected with        difference between infected and uninfected birds
MAV-2(N) as 12-day-old embryos or as 1-day-           was a 1.4-fold increase in relative spleen weight.
old hatched chicks suggests that the target cell      At 60 days, when the earliest evidence of neph-
for nephroblastoma is present at both stages of       roblastoma appeared in 3 of 12 birds, significant
differentiation. Line 15 x 7 chickens infected        increases were present in the weights of the
intravenously manifested the highest incidence        kidney, the liver, and the spleen. Chickens sac-
of osteopetrosis (50%). The incidence of osteo-       rificed during the interval of 90 to 130 days of
petrosis was comparable for line 15 x 7 chickens      age manifested increases in pancreas, bone, lung,
infected intraperitoneally and SPAFAS chick-          liver, and brain weights, and a decrease in thy-

                                                                                                             Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
ens infected intravenously (30 and 20%, respec-       mus weight relative to values for organs of un-
tively). However, SPAFAS chicks infected intra-       infected chickens. All 90- to 130-day-old infected
peritoneally manifested only 3% incidence of          birds possessed large nephroblastomas, and their
osteopetrosis.                                        mean body weight was half that of uninfected
   Influence of host age and route of infec-          chickens.
tion on the onset of nephroblastoma and                  Virus in organs of MAV-2(N)-infected
osteopetrosis. Chickens infected as embryos           chickens. Results of electron microscope stud-
by the intravenous route manifested an earlier        ies of nephroblastoma-bearing chickens infected
onset of nephroblastoma than those infected as        with AMV (20) and a nephroblastoma-inducing
hatched chicks by the intraperitoneal route (Ta-      isolate (51) indicate that virus particles are pres-
ble 2). This difference was especially evident in     ent in all organs with a large accumulation in
infected SPAFAS chicks; the average age of            the kidney. Consequently, organ extracts from
chickens dying or sacrificed with nephroblas-         MAV-2(N)-infected tumor-bearing chickens
toma was approximately one month younger for          were prepared to quantitate infectious virus.
intravenously infected embryos than for intra-        The results demonstrated that a correlation ex-
peritoneally infected hatched chickens. A slight      isted between the virus titer in the organs and
difference in the age of chickens dying or sacri-     the specificity of the virus for neoplasms of bone
ficed with osteopetrosis was apparent only with       and kidney (Fig. 1). Nephroblastoma, lung,
respect to strain, with SPAFAS chicks showing         bones, and pancreas show high virus titers rela-
a slightly earlier onset than did line 15 x 7         tive to that of serum, whereas liver, spleen,
chicks.                                               bursa, thymus, heart, and brain show progres-
   Contact transmission of MAV-2(N) infec-            sively lower relative titers. The statistical eval-
tion. A total of 34 uninfected SPAFAS and 15          uation of these results suffers from the low num-
 x 7 chickens were hatched and raised in the          ber of chickens involved and the difficulties of
same room with MAV-2(N)-infected chickens.            preparing extracts, particularly from bone.
Ten (29%) of the uninfected chickens demon-              Evidence of anemia in MAV-2(N)-in-
strated nephroblastoma. The mean age of con-          fected chickens. The association of anemia
tact-infected chickens manifesting nephroblas-        with infection by subgroup B avian leukosis
toma (111 ± 9 days) was slightly older than that      viruses has been well documented (3, 19, 37).
for experimentally infected chickens. None of         Thus, the PCV of MAV-2(N)-infected chickens
the contact-infected chickens demonstrated os-        relative to the PCV of uninfected chickens was
 teopetrosis.                                         monitored for 2 months after hatching. SPAFAS
   Growth rate of MAV-2(N)-infected chick-            chickens infected by the intravenous route did
 ens. To determine whether infection with MAV-        not manifest significant anemia. Only on day 11
 2(N) altered the growth rate of the chicken, the      (P < 0.02) was the decrease in PCV of infected
 body weight of intraperitoneally infected SPA-        chickens statistically significant with respect to
 FAS chicks was measured every third day for 2         the PCV of uninfected chickens. The PCV of
 months after hatching. Both infected and unin-        chickens bearing nephroblastomas (60 to 130
 fected chickens grew at a rate of 6.33 g per day.     days old) was also comparable to normal values.
 Therefore, no stunting of infected chickens was          Serum chemistry of MAV-2(N)-infected
 observed during this time period.                     chickens. Serum protein electrophoresis pro-
    Organ weights of MAV-2(N)-infected                 files of uninfected and MAV-2(N)-infected
 chickens. Organ weights of MAV-2(N)-infected          chickens revealed significant differences in al-
 chickens at 30, 60, and 90 to 130 days of age were    bumin and gamma globulin concentrations. At
 analyzed to determine whether systemic organ          30 and 60 days of age, serum albumin concentra-
 changes associated with MAV-2(N) infection            tion of infected birds was 69 and 83%, respec-
 could be detected. Organ weights were expressed       tively, of that of uninfected birds. Sera from
506     WATTS AND SMITH                                                                              INFECT. IMMUN.

                     TABLE 3. Relative organ weights of MA V-2(N)-infected chickens
                                                                 Relative organ wt
  Age   No. ex- Body wtb    p       -
 (days) amined'             tomac                                         Pan-         Bursa   Thymus Spleen       Bone
                                        Kidney   Liver   Lungs Brain      crease
          13 C     1.04;   Absent         1.04;     1.01;   1.06;
                                                             NS 0.97;
                                                                    NS 0.89;NS 1.02; NS 0.80;  1.37; 1.07;
                                                                                         NS
VOL. 27, 1980                                  AVIAN NEPHROBLASTOMA VIRUS PATHOLOGY                                      507
                                     TABLE 4. Serum neutralizing antibody titers
                                                                     Neutralizing antibody titerb
   Description of      Serum
      chicken'        (chicken       Serum        Serum      CEF          CEF         RSV           RSV     PR.RSVV
                         no.)        MAV-         MAV-       MAV-        MAV-        [MAV-      [MAV-                 RPV
                                     2(N)c        2(O)c      2(N)d       2(0)d       2(N)]          2(0)]      B
MAV-2(N)-im-             790           500           50      5,000         500       5,000            500      500    None
    munized              851            50            5        500          50         500             50       50    None
                        1584            50           50        500          50          50             50        5    None
                        1585           500          500      5,000         500         500             50      500    None
                        1586            50           50        500          50         500             50        5    None
MAV-2(0)-im-            1195           500           50        500          50          50             50       50    None
    munized             1215           500          500        500         500         500           500        50    None

                                                                                                                               Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
RPV-immunized           1197         None         None       None        None        None           None     None       500
                        1200         None         None       None        None        None           None     None       500
                        1201         None         None           5           5       None               5    None       500
MAV-2(N)-in-
     fected
  NE                   1076      None      None      None       None       None      None       None      None
  NO                   1078          5         5        50      None            5         5     None      None
  NE                   1079          5     None         50          5          50    None           5     None
  NE                   1082      None      None           5         5           5     None      None      None
  NE                   1084      None      None           5         5           5    None       None      None
  NE                   1086      None      None           5     None            5    None       None      None
  NE + OS              1087      None      None      None       None       None      None       None      None
  NO                   1093      None      None      None       None       None      None       None      None
  NE + OS              1098      None      None      None       None       None      None       None      None
  NE                   1898         50        50       500         50          50        50        50     None
  NE                   1915        500       500     5,000        500         500       500       500     None
  NE                   1443        500        50       500        500       5,000       500       500     None
   a Pathological manifestations of MAV-2(N)-infected chickens were scored as nephroblastoma (NE), nephro-
blastoma plus osteopetrosis (NE + OS), or normal (NO; no pathology at sacrifice).
   'Sera were analyzed for the presence of neutralizing antibody by plaque or focus neutralization assay as
described in the text. Neutralizing antibody titers are expressed as the reciprocal of the highest antiserum
dilution giving greater than 90% reduction in plaques or foci. The titer of sera having no detectable neutralizing
antibody is denoted "none."
   'Serum MAV-2(N) and serum MAV-2(O) denote virus present in infected chicken sera.
   d
     CEF MAV-2(N) and CEF MAV-2(O) denote virus obtained from infected chicken embryo fibroblast
cultures.

    TABLE 5. Relationship of serum neutralizing                  (Table 4). A similar pattern of type-specific neu-
  antibody titer to virus titer in MAV-2(N)-infected             tralizing antibody was not observed in a limited
                       chickens                                  number of MAV-2(0)-immunized chickens. The
                    Neutralizing anti-   Serum virus titer       neutralization titer for RSV pseudotypes corre-
   Chicken no."        body titer'           (PFU/ml)            lated generally with the neutralization titer for
       1431
508     WATTS AND SMITH                                                                INFECT. IMMUN.
increase in the gp85 content of MAV-2(0) rela-       avian nephroblastoma viruses. The DNV virus
tive to that of AMV and PR.RSV-C (48) was            of Lacour et al. (25) induced greater than 70%
paralleled by MAV-2(N). However, this in-            incidence of nephroblastoma with an onset of 45
creased gp85 content may be characteristic of        to 120 days in 1-day-old chicks injected intra-
many avian leukosis viruses: gels of RAV-O and       peritoneally. The latter virus strain also induced
RPV polypeptides likewise have shown consid-         some osteopetrosis. The virus of Ogura et al.
erable amounts of Coomassie blue staining ma-         (36), in contrast, induced no osteopetrosis when
terial migrating in the position of gp85 (E.         injected intraperitoneally into 1-day-old line 15
Schmidt and R. Smith, unpublished data). The         or Shaver chickens or intravenously into 11-day-
35S RNA of MAV-2(N) comigrated precisely in          old embryos. Approximately 55 to 75% of chick-
a single peak with that of MAV-2(0). Thus, the       ens infected with the latter virus strain devel-
genome of MAV-2(N), like that of MAV-2(0)            oped nephroblastomas with an onset from 60 to

                                                                                                          Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
(43), may be slightly larger than or the same size    120 days of age. Ogura et al. (36) also noted
as that of other nontransforming oncoviruses         contact transmission of nephroblastoma virus to
but is definitely smaller than that of avian sar-    uninfected hatchmates. A difference in the strain
coma viruses. The similarity in structural com-       of chickens used and the route of infection may
position of MAV-2(N) and MAV-2(0) may re-            be sufficient to explain some of the differences
flect a common evolutionary origin.                  in oncogenic spectrum of the original nephro-
   Results of the infection of chickens with         blastoma virus isolate and plaque-purified
plaque-purified MAV-2(N) both associate this         MAV-2(N). An analogous virus isolated from
virus with related avian oncoviruses and estab-       CEF transfected with AMV myeloblast DNA or
lish its distinctiveness with regard to the path-    line DNV DNA when injected intraperitoneally
ological features induced in infected chickens.       into 1-day-old chicks also induced nephroblas-
By gross and histological examination, nephro-       tomas with a similar onset (16).
blastomas induced by cloned MAV-2(N) were                Variation in the incidence of osteopetrosis in
comparable to those induced: by AMV, BAI              SPAFAS chickens with the route of MAV-2(N)
strain A (20, 21); by transplanted homogenates        infection (Table 2) seems analogous to the vari-
of AMV-induced nephroblastomas (50); by avian         ation in the incidence of erythroblastosis versus
nephroblastomas virus line DNV (26); by the           sarcomas upon injection of avian erythroblasto-
original subgroup B nephroblastoma-inducing           sis virus strain ES4 by the intravenous route
virus isolated by transfection (36); and by MAV-      versus the intramuscular route (18). That is, if
2(0). The only neoplasm induced by MAV-2(N)          MAV-2(N) has the potential to induce both
other than nephroblastoma was an osteopetrosis       nephroblastoma and osteopetrosis, infection by
similar or identical to that induced by MAV-         the intraperitoneal route may promote the ap-
2(0).                                                pearance of nephroblastoma over osteopetrosis.
   Whether MAV-2(N) and MAV-2(0) are mon-            Such an enhancement must be related only to
opotent or pluripotent with respect to oncogenic     incidence and not to onset, because both SPA-
spectrum is an unresolved question. The wide          FAS and line 15 x 7 chickens infected with
oncogenic spectrum of AMV results from the           MAV-2(N) demonstrated an earlier onset of
constitution of standard strains of AMV with         nephroblastoma when infected intravenously as
defective leukemogenic viruses and helper oste-      embryos than intraperitoneally as hatched
petrosis and nephroblastoma viruses (45). How-       chicks (Table 2).
ever, isolation of MAV-2(N) and MAV-2(0) by              The pathological characteristics associated
plaque formation presumes their nondefective-        with MAV-2(N) infection are of significance in
ness for replication. Thus, the overlap in onco-     considering the course of disease and in compar-
genic spectrum of MAV-2(N) and MAV-2(0) is           ing the biological effects of MAV-2(N) and
a problem which likely cannot be explained by        MAV-2(0). These characteristics may be related
contamination of cloned virus stocks with the        to the process of oncogenesis or may only reflect
other strain or by virus defectiveness. However,     virus infection and replication. Although osteo-
MAV-2(N) and MAV-2(0) may be pluripotent             petrotic chickens grew at a rate 26% of the rate
virus strains having been selected for specific      of uninfected chickens (3), MAV-2(N)-infected
oncogenicity by their isolation. However, com-       chickens through 2 months of age grew at a rate
parative nucleic acid studies have indicated that    comparable to that of uninfected chickens. How-
the genomes of MAV-2(N) and MAV-2(0) are             ever, the mean body weight of MAV-2(N)-in-
indeed different (S. Watts and R. Smith, manu-       fected chickens sacrificed at 90 to 130 days of
script in preparation).                              age was 54% of that of uninfected chickens (Ta-
  The incidence and onset of nephroblastoma          ble 3). The latter result may reflect debilitation
induced by MAV-2(N) are entirely comparable          resulting from tumor mass or stunting due to
to those indexes found with infection by other       decreased growth rate.
VOL. 27, 1980                            AVIAN NEPHROBLASTOMA VIRUS PATHOLOGY                       509
   Organs of 4-week-old osteopetrotic chicks          fected with nephroblastoma-inducing viruses
demonstrated a decrease in lymphoid organ             (20, 51). Results of electron microscopic studies
weight and increases in the weights of heart,         have indicated that the number of budding virus
alimentary tract, bones, liver, kidneys, and brain    particles was greatest in the target organs, bone,
(3). However, MAV-2(N)-infected chicks                and kidney after infection with a nephroblas-
showed an increase in spleen weight at 1 and 2        toma virus (51) or with MAV-2(0) (9, 52; E.
months of age (Table 3). At greater than 3            Schmidt and R. Smith, unpublished data). The
months of age, MAV-2(N)-infected chickens             latter phenomenon seems to be confirmed by
demonstrated increases in the relative weights        the titration of infectious virus in extracts of
of liver, lungs, brain, pancreas, and bones as well   organs of MAV-2(N)-infected chickens. The pri-
as a large decrease in thymus weight. In MAV-         mary lymphoid organs, the bursa and thymus,

                                                                                                            Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
2(0)-infected chicks, the decrease in lymphoid        contained relatively little virus compared with
organ weight has been associated with immu-           the nephroblastoma, bone, lung, and pancreas
nosuppression (47), whereas the increase in           (Fig. 1). These results suggest that the replica-
weight of other organs was attributed to edema        tion of MAV-2(N) in cells of the lymphoid or-
(3). Specific viral proteins have been associated     gans is limited. The present study did not ad-
with lymphoid organ atrophy and immuno-               dress the question of whether MAV-2(N) was
suppression upon infection of kittens with feline     capable of transforming cells of the bursa. All
leukemia virus (1, 28). The consequences of           infected chickens died or were sacrificed (129
lymphoid organ atrophy in MAV-2(N)-infected           days [Table 2]) before sufficient time had
chickens have not been examined but edema             elapsed for the manifestation of lymphoid leu-
may be involved in the increase in weight of          kosis. Nevertheless, elevated virus titers in the
some organs of MAV-2(N)-infected chickens.            nephroblastoma and bone suggest that replica-
However, an apparent increase in relative organ       tion may play a role in organ-specific oncogen-
weight may result from a decrease in body             esis.
weight due to debilitation and wasting of the             The presence of neutralizing antibody and
nephroblastoma-bearing chicken.                       nephroblastoma in certain infected chickens
    Unlike chickens infected with MAV-2(0) (37),       (Tables 4 and 5) requires comment. First, the
chicken embryos infected intravenously with           presence of neutralizing antibody indicates that
 MAV-2(N) did not develop significant anemia          the birds were infected under conditions which
after hatching. However, other investigations          did not permit tolerance to the virus. It is pos-
have indicated that some chickens infected in-         sible that infection of the day-old chick by the
 travenously at 8 to 9 days of age with MAV-2(N)       intraperitoneal route permitted the infection of
 developed a transient anemia (R. Paterson and         certain target cells, such as those of the mesen-
 R. Smith, unpublished data). Thus, the associ-        chymal rest (5), and once infected, these cells
 ation of anemia with MAV-2(N) infection seems         were destined to become transformed, despite
 to be ambiguous at this time. Anemia induced          the presence of antibody. Second, it seems clear
 by MAV-2(0) was accompanied by a thrombo-             that the presence of viremia and the absence of
 cytopenia (37). That MAV-2(N)-infected chick-         neutralizing antibody did not accelerate the ap-
 ens had petechial hemorrhages might suggest a         pearance of nephroblastoma. It seems therefore
 blood disorder promoting capillary fragility.         likely that there is little recruitment of new
 Petechial hemorrhages may therefore lead to           target cells by virus after the initial infection.
 the excess of cannabilism observed in MAV-            This is in contrast to the situation in MAV-2(0)
 2(N)-infected chickens.                               infection, in which administration of neutralizing
    The changes in albumin and gamma globulin          antibody leads to an arrest of disease (R. E.
 levels detected in the serum of MAV-2(N)-in-          Smith and J. Ivanyi, manuscript in preparation),
 fected birds parallel those seen in MAV-2(0)-         suggesting a continuous recruitment of new tar-
 infected chickens (3). Sanders et al. (39) also       get cells by MAV-2(0). It is possible that a
 noted an increase in a y globulin-associated com-     limited number of transformable target cells ex-
 ponent and a decrease in the albumin/total glob-       ists for MAV-2(N), and that these cells remain
 ulin (A/G) ratio for sera from leukosis-affected       susceptible to virus transformation for a rela-
 chickens relative to uninfected chickens. Also,        tively long time during development. Further
 the increased serum gamma globulin level may           experiments are required to elucidate this point,
  also be a consequence of chronic virus infection.     particularly those employing the infection of
     The virus titer in organ extracts of nephro-       embryos with MAV-2(N) followed by the admin-
  blastoma-bearing chickens was greater than that       istration of neutralizing antibody.
  in serum (Fig. 1). The latter observation corre-         Neutralizing antibody titers reported for
  lates with the finding of budding virus by elec-      MAV-2(N)-infected chickens indicate that the
  tron microscopy in all organs of chickens in-         presence of nephroblastoma was not always as-
510    WATTS AND SMITH                                                                          INFECT. IMMUN.
sociated with virus-specific immunoglobulin.          of adults with a high dose of virus will lead to
Furthermore, preliminary results indicated that       infection and ultimately to transformation of
treatment of chickens with cyclophosphamide           target cells, if present, in spite of the develop-
did not prevent nephroblastoma induction by           ment of neutralizing antibodies. Second, the ge-
MAV-2(N). Nowygrod et al. (34) likewise found         netics of disease induction could be investigated.
that growth of AMV-induced nephroblastomas            Lymphoid leukosis induction is influenced by at
did not require the presence of circulating anti-     least one gene other than that coding for a
body. However, the level of serum antibody            surface receptor for virus (12). Since disease
demonstrated a striking inverse correlation with      induction in nephroblastoma is relatively rapid
the level of serum virus in MAV-2(N)-infected         (compared with lymphoid leukosis) it would be
chickens. Thus, the antibody response in in-          of interest to determine whether several inbred

                                                                                                                       Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
fected chickens may play a significant role in        lines of chickens differ in susceptibility to neph-
limiting viremia, even though not related to          roblastoma induction. Genetic analysis could im-
nephroblastoma induction. As shown with RSV-          plicate the same genetic loci for lymphoid leu-
induced sarcomas, virus immunity may have no          kosis and nephroblastoma. Third, the role of
relationship to or may enhance tumor growth           immunological competence could be addressed
(29). Furthermore, virus-antibody complexes           with the nephroblastoma model. Infection of
may be infectious (40).                               bursectomized adult chickens could be used to
   Moreover, as shown by the titration of serum       investigate the role of antibody in the disease
antibody to MAV-2(N) and related viruses (Ta-         process. If nephroblastomas are observed in bur-
ble 4), in some chickens the humoral immune           sectomized chickens, the role of antibody in the
response detectable by neutralization assay           disease process will be eliminated. A preliminary
manifested specificity for the infecting virus. As    experiment with a limited number of cyclophos-
shown by competition radioimmunoassay (48),           phamide-treated chickens indicated that these
MAV-2(0) virus or MAV-2(0) gp85 isolated              animals would develop nephroblastomas. How-
from CEF cultures and MAV-2(0) from the               ever, more complete experiments are planned,
serum of osteopetrotic chickens showed different      in which chicks which have been treated with
antigenic reactivities. In correlation, the neu-      both testosterone and cyclophosphamide will be
tralizing activity of some of the MAV-2(N)-in-        employed, and treated chicks will be tested for
fected or -immunized chicken sera characterized       agammaglobulinemia before administration of
in Table 4 also demonstrated preference for CEF       virus. If no tumors are observed in bursecto-
versus serum virus antigens. Virus neutralization     mized chickens, passive antibody administration
patterns of some chicken sera also indicated a        to infected chickens might lead to the appear-
close antigenic relatedness between MAV-2(N)          ance of nephroblastomas if antigen-antibody
and MAV-2(0)-closer than the relatedness be-          complexes are involved in the disease process.
tween either or these two viruses and PR.RSV-         The role of lymphocytes can be investigated by
B. Lack of neutralization of RPV with sera from       the adoptive transfer of immune and nonim-
chickens infected or immunized with subgroup          mune lymphocytes from histocompatible do-
B virus reflected the subgroup-specific nature of     nors. The experiments proposed should allow
virus antigenicity associated with the envelope       definitive answers to several interesting biologi-
glycoprotein. Likewise, the type-specific reactiv-    cal questions.
ities of these sera also may be associated with
the glycoprotein determinants of the virus (23).                       ACKNOWLEDGMENTS
   The present study presents a description of           The technical contributions of T. Wheeler and S. Nebes
the pathological manifestations of chickens in-       provided indispensable support for this investigation. A. J.
                                                      Banes gave advice and support during the inception and
fected with an avian nephroblastoma virus. The        analysis of these experiments. We wish to especially thank
results of this investigation indicate that the       Heinz Bauer for generously providing the original isolate of
model system employed would be useful to ad-          nephroblastoma virus.
dress several fundamental questions of RNA               This research was supported by Public Health Service
                                                      grants R01-CA-12323 and P01-CA-14236 from the National
tumor virus pathogenesis. First, the number and       Cancer Institute. S.L.W. was a recipient of a National Science
persistence of target cells in the host during        Foundation Predoctoral Fellowship HES-7422381.
development could be investigated by infecting                          LITERATURE CITED
both embryos and adult animals. The number             1. Anderson, L. J., W. F. H. Jarrett, 0. Jarrett, and H.
of target cells could be estimated by microscopic           M. Laird. 1971. Feline leukemia virus infection of kit-
enumeration of transformed foci present in the              tens: mortality associated with atrophy of the thymus
kidney at an early stage of tumor formation (i.e.,         and lymphoid depletion. J. Nati. Cancer Inst. 47:807-
60 days postinfection) and observation of in-              817.
                                                      2. Baluda, M. A., and P. P. Jamieson. 1961. In vivo
fected animals for nephroblastomas for a period            infectivity studies with avian myeloblastosis virus. Vi-
of at least 6 months. It is proposed that injection        rology 14:33-45.
VOL. 27, 1980                                          AVIAN NEPHROBLASTOMA VIRUS PATHOLOGY                                      511
  3. Banes, A. J., and R. E. Smith. 1977. Biological charac-                 from standard avian myeloblastosis virus. J. Virol. 15:
        terization of avian osteopetrosis. Infect. Immun. 16:                906-912.
       876-884.                                                       23. Ishizaki, R., and P. K. Vogt. 1966. Immunological rela-
 4. Beard, J. W. 1963. Avian virus growths and their etiologic               tionships among envelope antigens of avian tumor vi-
       agents. Adv. Cancer Res. 7:1-127.                                     ruses. Virology 30:375-387.
  5. Beard, J. W., J. F. Chabot, D. Beard, U. Heine, and              24. Jones, T. L. 1952. Embryonal nephroma in a dog. Can. J.
        G. E. Houts. 1976. Renal neoplastic response to leu-                 Comp. Med. 16:153-165.
        kosis virus strains BAI A (avian myeloblastosis virus)        25. Lacour, F., E. Delain, R. Gerard-Marchant, and 0.
        and MC29. Cancer Res. 36:339-353.                                    Weiler. 1970. Dysembryome nephroblastique "lign6e
  6. Beaudreau, G. S., C. Becker, R. A. Bonar, A. M.                         DNV" du poulet. Observations de 10 generations de
       Wallbank, D. Beard, and J. W. Beard. 1960. Virus                      transmission experimentale. C. R. Acad. Sci. Paris Serie
       of avian myeloblastosis. XIV. Neoplastic response of                  D 271:141-144.
       normal chicken bone marrow treated with the virus in           26. Lacour, F., 0. Weiler, R. Gerard-Marchant, and E.
       tissue culture. J. Natl. Cancer Inst. 24:395-415.                     Delain. 1970. Transformation et differenciation du tissu

                                                                                                                                         Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
  7. Bell, E. T., and A. T. Henrici. 1916. Renal tumors in the               renal par un virus oncogene a RNA. Bull. Cancer 57:
       rabbit. J. Cancer Res. 1:157-167.                                     335-344.
  8. Bolognesi, D. P., and H. Bauer. 1970. Polypeptides of            27. Lowry, 0. H., N. J. Rosebrough, A. L Farr, and R. J.
        avian RNA tumor viruses. I. Isolation and physical and               Randall. 1951. Protein measurement with the Folin
       chemical analysis. Virology 42:1097-1112.                             phenol reagent. J. Biol. Chem. 193:265-275.
  9. Boyde, A., A. J. Banes, R. M. Dillaman. and G. L                 28. Mathes, L E., R. G. Olsen, L. C. Hebebrand, E. A.
       Mechanic. 1979. A morphological study of an avian                     Hoover, and J. P. Schaller. 1978. Abrogation of lym-
        bone disorder caused by myeloblastosis-associated vi-                phocyte blastogenesis by a feline leukemia virus protein.
       rus. Metab. Bone Dis. Rel. Res. 1:235-242.                            Nature (London) 274:687-689.
10. Burmester, B. R., M. A. Gross, W. G. Walter, and A.               29. McBride, R. A., D. H. Watanabe, and L. W. Schier-
        K. Fontes. 1959. Pathogenicity of a viral strain (RPL                man. 1977. Rous sarcomas in chickens: enhanced
        12) causing avian visceral lymphomatosis and related                 growth coexisting with concomitant immunity. Science
        neoplasms. II. Host-virus interrelations affecting re-               197:1079-1082.
       sponse. J. Natl. Cancer Inst. 22:103-127.                      30. Moscovici, C. 1975. Leukemic transformation with avian
11. Collett, M. S., and A. J. Faras. 1977. In vitro transcrip-               myeloblastosis virus: present status. Curr. Top. Micro-
        tion of the avian oncornavirus genome by the RNA-                    biol. Immunol. 71:79-101.
        directed DNA polymerase: analysis of DNA transcripts          31. Moscovici, C., and P. K. Vogt. 1968. Effects of genetic
        synthesized in reconstructed enzymatic reactions. J.                 cellular resistance on cell transformation and virus rep-
        Virol. 22:86-96.                                                     lication in chicken hematopoietic cell cultures infected
12. Crittenden, L. B. 1975. Two levels of resistance to lymph-               with avian myeloblastosis virus (BAI.A). Virology 35:
        oid leukosis. Avian Dis. 19:281-292.                                 487-497.
13. Faras, A. J., J. M. Taylor, J. P. McDonnell, W. E.                32. Moscovici, C., and M. Zanetti. 1970. Studies on single
        Levinson, and J. M. Bishop. 1972. Purification and                   foci of hematopoietic cells transformed by avian mye-
        characterization of the deoxyribonucleic acid polymer-               loblastosis virus. Virology 42:61-67.
        ase associated with Rous sarcoma virus. Biochemistry          33. Nicholson, G. W. 1931. An embryonic tumour of the
        11:2334-2342.                                                        kidney in a foetus. J. Pathol. Bacteriol. 34:711-730.
14. Feldman, W. H. 1932. Neoplasms of domesticated ani-               34. Nowygrod, R., D. E. R. Sutherland, R. J. Howard,
        mals, p. 357-372. W. B. Saunders and Co., Philadelphia.              and J. S. Najarian. 1977. Tumor growth in the absence
15. Floyd, R. W., M. P. Stone, and W. K. Joklik. 1974.                       of circulating antibodies. J. Surg. Res. 22:660-666.
        Separation of single-stranded ribonucleic acids by acryl-     35. Ogura, H., R. R. Friis, and H. Bauer. 1974. Isolation of
        amide-agarose-urea gel electrophoresis. Anal. Biochem.               infectious DNA from avian myeloblastosis virus trans-
        59:599-609.                                                          formed leukemic cells. Z. Naturforsch. Teil C 29:437-
16. Fourcade, A., T. Huynh, and F. Lacour. 1974. Trans-                      441.
        fection of chicken embryo cells with DNA extracted            36. Ogura, H., H. Gelderblom, and H. Bauer. 1974. Isola-
        from avian virus-producing neoplastic cells. J. Virol. 14:           tion of avian nephroblastoma virus from avian myelo-
        407-411.                                                             blastosis virus by the infectious DNA technique. Inter-
17. Graf, T. 1972. A plaque assay for avian RNA tumor                        virology 4:69-76.
        viruses. Virology 50:567-578.                                 37. Paterson, R. W., and R. E. Smith. 1978. Characteriza-
18. Graf, T., D. Fink, H. Beug, and B. Royer-Pokora.                         tion of the anemia induced by an avian osteopetrosis
        1977. Oncornavirus-induced sarcoma formation ob-                     virus. Infect. Immun. 22:891-900.
        scured by rapid development of lethal leukemia. Cancer        38. Plummer, P. J. G. 1951. A survey of sixty tumours from
        Res. 37:59-63.                                                       domesticated animals. Can. J. Comp. Med. Vet. Sci. 15:
19. Graf, T., B. Royer-Pokora, G. E. Schubert, and H.                        231-253.
        Beug. 1976. Evidence for the multiple oncogenic poten-        39. Sanders, E., L. F. Huddleson, and P. J. Schaible. 1944.
        tial of cloned leukemia virus: in vitro and in vivo studies          An electrophoretic study of serum and plasma from
        with avian erythroblastosis virus. Virology 71:423-433.              normal and leucosis-affected chickens. J. Biol. Chem.
20. Heine, U., G. de The, H. Ishiguro, J. R. Sommer, D.                       155:469-481.
        Beard, and J. W. Beard. 1962. Multiplicity of cell            40. Schlesinger, M. J. 1976. Formation of an infectious virus-
        response to the BAI strain A (myeloblastosis) avian                  antibody complex with Rous sarcoma virus and anti-
        tumor virus. II. Nephroblastoma (Wilms' tumor): ultra-               bodies directed against the major virus glycoprotein. J.
        structure. J. Natl. Cancer Inst. 29:41-105.                          Virol. 17:1063-1067.
21. Ishiguro, H., D. Beard, J. R. Sommer, U. Heine, G.                41. Smith, R. E. 1978. Large scale growth of Rous sarcoma
        de The, and J. W. Beard. 1962. Multiplicity of cell                  virus. In W. B. Jacoby and I. H. Pastan (ed.) Methods
        response to the BAI strain A (myeloblastosis) avian                  Enzymol. Vol. 58, p. 393-403. Academic Press Inc., New
         tumor virus. I. Nephroblastoma (Wilms' tumor): gross                 York.
         and microscopic pathology. J. Natl. Cancer Inst. 29:1-       42. Smith, R. E., and E. H. Bernstein. 1973. Production and
         39.                                                                  purification of large amounts of Rous sarcoma virus.
 22. Ishizaki, R., A. J. Langlois, and D. P. Bolognesi. 1975.                 Appl. Microbiol. 25:346-353.
         Isolation of two subgroup-specific leukemogenic viruses       43. Smith, R. E., L. J. Davids, and P. E. Neiman. 1976.
512        WATTS AND SMITH                                                                                       INFECT. IMMUN.
        Comparison of an avian osteopetrosis virus with an                 avian osteopetrosis virus: effect of host cell on antigenic
        avian lymphomatosis virus by RNA-DNA hybridiza-                    reactivity. Virology 90:80-89.
        tion. J. Virol. 17:160-167.                                49.   Vogt, P. K. 1969. Focus assay of Rous sarcoma virus, p.
44.   Smith, R. E., and F. Kozoman. 1973. Device for auto-                 198. In K. Habel and N. P. Salzman (ed.), Fundamental
        matic rapid harvest of roller culture supernatant fluid.           techniques of virology. Academic Press Inc., New York.
        Apple. Microbiol. 25:1008-1010.                                    N. Y.
45.   Smith, R. E., and C. Moscovici. 1969. The oncogenic          50.   Walter, W. G., B. R. Burmester, and C. H. Cun-
        effects of nontransforming viruses from avian myelo-               ningham. 1962. Studies on the transmission and pa-
        blastosis virus. Cancer Res. 29:1356-1366.                         thology of a viral-induced avian nephroblastoma (em-
46.   Smith, R. E., and K. Quade. 1976. Production of large                bryonal nephroma). Avian Dis. 6:455-477.
        quantities of 32P-labeled RNA tumor virus nucleic acid.    51.   Weiler, O., E. Delain, and F. Lacour. 1971. Studies on
        Anal. Biochem. 70:354-358.                                         a viral nephroblastic nephroblastoma of the chicken: an
47.   Smith, R. E., and L. J. Van Eldik. 1978. Characteriza-               electron microscope comparison of the sequence of de-
        tion of the immunosuppression accompanying virus-                  velopment of the visions in different organs. Eur. J.

                                                                                                                                         Downloaded from http://iai.asm.org/ on January 13, 2021 by guest
        induced avian osteopetrosis. Infect. Immun. 22:452-461.            Cancer 7:491-494.
48.   Van Eldik, L. J., and R. E. Smith. 1978. Isolation and       52.   Young, D. 1966. Osteopetrosis virus growth in the kidney
        characterization of the envelope glycoprotein of an                of infected chickens. Eur. J. Cancer 2:13-18.
You can also read