You are on page 1of 14

Adenoviruses Secondary article

S Jane Flint, Princeton University, Princeton, New Jersey, USA Article Contents
. Introduction
Adenoviruses are nonenveloped viruses with double-stranded DNA genomes that are . Classification
generally associated with relatively mild, self-limiting diseases in humans. They rely on . Structure
cellular systems for expression of viral genetic information during reproduction in . Replication
permissive cells and can transform normal cells in culture; some are tumorigenic in rodents. . Epidemiology
. Clinical Features

Introduction . Viral Oncogenesis

Adenoviruses, which appear to be restricted to warm-


blooded animals, are defined by their linear, double- end result of infection is the production of large quantities
stranded deoxyribonucleic acid (DNA) genomes and the of the viral genome and protein components of virions for
characteristic size and structure of virions. Human assembly of progeny virions and death of the host cell.
members of this family are important causes of respiratory The processes necessary for successful adenovirus
disease and gastroenteritis in children and of conjunctivitis reproduction, from initial attachment to maturation and
in children and adults, but in general they are not highly release of progeny virions, can be outlined in some detail.
pathogenic. Nevertheless, adenoviruses have received Nevertheless, many questions remain about the molecular
considerable attention, in part because this family includes mechanisms underlying important regulatory circuits
the first human viruses shown to be tumorigenic in established by viral proteins and the production of progeny
experimental animals. Most adenoviruses can convert virions. Within the past decade, there has been great
normal, nontumorigenic rodent cells in culture to trans- resurgence of interest in adenoviruses, because of their
formed cells with tumorigenic potential. Investigation of experimental use as vectors in gene therapy or in anticancer
the molecular basis of adenovirus transformation spurred therapies. Broad cell tropism, ability to infect highly
development of a general paradigm for transformation by differentiated cells, and the ease with which the viral
small DNA tumour viruses such as the Simian virus 40 genome can be manipulated are some of the properties that
(SV40) and polyoma papovaviruses. More importantly make these viruses attractive as vectors for therapeutic
perhaps, such studies led to much improved understanding purposes. Many specific protocols are now being tested,
of both the mechanisms of action of the products of cellular and much effort is being devoted to the development of
tumour suppressor genes and the circuits that govern adenoviral vectors with optimal features. One dividend has
orderly progression of normal cells through the cell cycle, been the greatly improved understanding of crucial
and hence their growth and proliferation. reactions in the infectious cycle, notably the mechanisms
Like all viruses, adenoviruses are molecular parasites of adenovirus attachment and entry into host cells.
that exploit cellular biosynthetic systems for their replica-
tion. In permissive host cells, adenoviruses direct synthesis
of viral proteins that replicate the viral DNA genome, but Classification
rely on cellular transcription, pre-mRNA (messenger
ribonucleic acid) processing and translation systems for Adenoviruses were named in recognition of the isolation of
expression of viral genetic information. Consequently, the prototype strain, Human adenovirus 2 (Ad2), from
much fundamental information about mechanisms that young children’s adenoids placed in culture (Rowe et al.,
mediate and regulate cellular gene expression has come 1953). Their definitive properties include linear, double-
from investigation of adenoviral reproduction, including stranded DNA genomes of 30 000–45 000 bp and none-
the splicing of pre-mRNA that is a characteristic feature of nveloped particles, 80–110 nm in diameter, which exhibit a
eukaryotic cells (Berget et al., 1977; Chow et al., 1977). striking icosahedral appearance and carry one or two fibres
Adenoviruses are not, however, merely passive competi- projecting from each vertex (Figure 1a). Many viruses with
tors for access to these cellular systems, for the adenoviral these properties have been isolated from humans, other
genome encodes a number of regulatory proteins that primates, domesticated mammals, rodents, marsupials
ensure orderly and efficient expression of viral coding and birds. In 1976, the International Committee on
sequences. The strictly defined sequence in which viral Taxonomy of Viruses recognized this group of viruses as
genes are expressed is determined primarily by regulation the family Adenoviridae, comprising the genera Mastade-
of transcription, while posttranscriptional mechanisms novirus and Aviadenovirus.
induce inhibition of cellular mRNA production and These genera were defined originally on the basis of the
protein synthesis as the infectious cycle progresses. The taxonomic class of the virus host (Mammalia and Aves,

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 1


Adenoviruses

Figure 1 Structure of adenovirus particles. (a) Surface view of the Ad2 virion obtained by cryoelectron microscopy and image reconstruction. The fibres
project some 33 nm, but only the portions proximal to the virion surface are seen, because these structures are bent. This view, which is orientated
along an icosahedral axis of 3-fold rotational symmetry, has a nominal resolution of 30 Å. From Stewart PL et al. (1991) Cell 67: 145–154, with
permission. (b) Schematic section through the virion, illustrating the locations of the virion proteins and DNA genome. The organization of the capsid
proteins shown is based on biochemical and structural studies. However, protein VIII has not been localized, even though it is known to be internal, and is
shown associated with hexons because it is believed to stabilize the capsid. In the core, the viral DNA is shown associated with protein VII (dashed lines), the
major DNA-binding protein of the virion. There have been reports of organization of the DNA into spherical domains, but in the absence of more detailed
structural information, no specific structure is shown for the core. Adapted from Flint SJ et al. (2000) Principles of Virology: Molecular Biology, Pathogenesis
and Control, ASM Press, Washington, DC, with permission.

respectively), and the presence of group-specific antigens in and biochemical properties. More recent phylogenetic
the hexon, the major structural protein of the virion; analyses of specific coding or control sequences of
however, mammalian and avian adenoviruses can also be mastadenoviral genomes have confirmed this classification
distinguished by the sizes, sequences and organization of scheme (Bailey and Mautner, 1994).
their genomes. Those of the former are significantly smaller
than those of the latter, 35 937 bp for Ad2 and 43 804 bp for
Chicken embryo lethal orphan (CELO) virus, respectively.
Moreover, the CELO virus genome lacks certain coding Structure
sequences found in all mastadenoviral DNAs examined
and includes long sequences at either end with no Electron microscopy of negatively stained adenovirus
counterparts in the genomes of Mastadenovirus. particles established that the capsid is built from 252
More than 120 adenovirus serotypes have been distin- structural units, 12 pentons located at the axes of 5-fold
guished largely by immunological tests. Individual ser- rotational symmetry (vertices) of the icosahedral virion
otypes can be distinguished by genomic differences, now and 240 hexons that form the remainder of the protein shell
often detected by DNA sequencing. Such molecular (Figure 1). These structural units are formed from three viral
approaches also permit the identification of variants of proteins (II, III and IV), but virions contain nine additional
individual serotypes, often called genome types. Most proteins that have been ascribed specific structural or
attention has been paid to human adenoviruses and their functional roles (Table 2). The current model of these
classification into subgroups (subgenera). Currently, the complex and large particles (some 150  106 Da in the case
Adenoviridae includes 49 human serotypes (Ad1–Ad49). of Ad2) (Figure 1b) is based on biochemical studies and
These viruses have been classified into subgroups using cryoelectron microscopy and image reconstruction of
biological properties, structural or biochemical features, subgroup C human adenoviruses.
and the relatedness of their genomes (Table 1). There is Most Mastadenovirus species carry a single fibre, with a
remarkably good agreement among these disparate fixed, serotype-specific length (Table 1), projecting from the
criteria. The six human adenovirus subgroups, A–F, were penton at each vertex of the virion (Figure 1); however, some
defined by the degrees of genome relatedness, but all carry both long and short fibres (one at each vertex)
members of each subgroup also share biological, structural (subgroup F in Table 1), while two fibres project from each
penton base of all but one avian adenoviruses. Adenoviral

2 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Table 1 Some properties of human adenoviruses
Fibre % Relatedness Oncogenicity in
Subgroup Serotypes lengths a (nm) of DNAb newborn rodents Haemagglutination Most common disease syndromes
ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net

A 12, 18, 31 28–31 48–69 High Little or none (type IV) Diarrhoea; most frequently
(8–20) isolated from stool specimens
B 3, 7, 11, 14, 16, 9–11 89–94 Moderate Complete agglutination of Upper and lower respiratory
21, 34, 35 (9–20) monkey erythrocyctes infections, e.g. bronchitis,
(type I) pneumonia (especially Ad7);
acute respiratory disease;
pharyngoconjunctival fever
C 1, 2, 5, 6 23–31 99-100 None Partial agglutination of rat Upper and lower respiratory
(10–16) erythrocytes (type III) infections in young children;
endemic
D 8–10, 13, 15, 17, 12–13 95–99 Nonec Complete agglutination of Epidemic keratoconjunctivis
19, 20, 22–30, 32, (4–17) rat erythrocytes (type II) (Ad8, 19 and 37); conjunctivitis
33, 36–39, 42–49
E 4 17 NA None Type III Acute respiratory disease;
(4–23) pharyngoconjunctival fever
F 40, 41 33 62–69 None Type III Gastroenteritis
22 (15–22)
NA, not applicable.
a
Values for the length of the fibre projecting beyond the penton base estimated from electron micrography of purified virions or pentons. For subgroup F (Ad40), the total lengths of the two
fibres including the portion within the penton base were measured. The values listed were calculated by subtracting the length of the latter portion (~4 nm) from the reported values.
b
The values shown were obtained by liquid hybridization between pairs of the viral DNA genomes listed in column 2. Those in parentheses are from hybridization between the DNAs of a
serotype within the subgroup and one belonging to a different subgroup.
c
With the exception of Ad9, which induces oestrogen-dependent mammary tumours in female rats of a certain strain.

Adenoviruses
3
Adenoviruses

Table 2 Adenovirus type 2 virion proteins


Molecular mass
Proteina (kDa) b Number per virionc Location and function(s)
II 110 720 Hexons (trimers of II); major structural unit of the capsid
III 63 60 Penton bases (pentamers of III); structural units at axes of
5-fold rotational symmetry; anchor fibres; bind to cell
surface integrins to induce internalization of particles and
penetration of endosomal membrane
IIIa 63 60 Monomers span from exterior to interior of capsid at the
edges between hexon faces; stabilize capsid
IV 62 36 Fibres (trimers of IV) projecting from penton bases; distal
knob carries binding sites for cellular receptor
V 42 157 ± 1 Core, probably on outer surface; contacts protein VI;
packaging of DNA genome
Terminal protein (TP) 38 2 Core, monomer covalently linked to the 5′ end of each
strand of the genome; protease cleavage product of the
protein primer for viral DNA synthesis
L3 protease 23 ~20 Cleaves precursors to proteins IIIa, VI, VII, VIII, µ and TP
following assembly to convert immature to infectious
virions; required for uncoating
VI 22 34.2 ± 4 Hexamers associated with peripentonal hexons; stabilizes
capsid; contacts components of the core
VII 19 833 ± 19 Core, bound to viral DNA; major DNA packaging protein
VIII 15 127 ± 3 Internal surface of capsid shell; stabilization of capsid?
IX 14 240 Trimers on exterior surface of the capsid, with monomers
extending along hexon–hexon interfaces in the centre of
each face of the icosahedral particle; stabilizes capsid
µ 2 125–160 Core; packaging of the genome
a
Virion proteins were originally named with Roman numerals on the basis of their separation by electrophoresis in SDS-polyacrylamide gels. In
addition to those listed, virions contain a number of small polypeptides produced when structural protein precursors are processed by the L3
protease.
b
Calculated from predicted amino acid sequence, and rounded to the nearest kilodalton.
c
Absolute numbers per virion are from the current model of Ad2 particles obtained by three-dimensional reconstruction of images collected by
cryoelectron microscopy. All others were determined by biochemical methods.

fibres are responsible for the initial attachment of particles crystal structure, the Ad5 fibre knob resembles a three-
to host cells, and fibre length may be an important bladed propeller, one blade contributed by each monomer.
determinant of attachment and internalization mechan- The location of the structural features necessary for CAR
isms. All fibres examined exhibit a conserved organization: binding suggests that the fibre carries three binding sites for
a capsid-distal, globular knob is separated from a ‘tail’ the viral receptor.
domain by a long, thin shaft formed from the central The penton base anchors the fibre to the particle, must
domains of the three protein IV monomers (Table 2). In bind stably to the five surrounding hexons (Figure 1) and is
each monomer, this central segment contains multiple required for internalization of virions. In side view, it
copies of a pseudorepeat of 15 amino acids, defined by the appears as a hollow bucket with a central hole into which
presence of conserved amino acids at specific positions, the tail of the fibre is inserted. On its outer surface, each of
which form a triple spiral structure in the trimer. The highly the protein III subunits (Table 2) possesses a groove that
conserved tail domain and the knob are responsible for could accommodate a b-barrel motif of the hexon subunits
binding of the fibre to the penton base and to the cellular (see below). During adenovirus entry, the penton base
receptor for attachment, respectively. Most human ser- binds to specific integrins on the host cell surface, an
otypes bind to the same cellular protein, the coxsackievirus interaction that requires the sequence Arg-Gly-Asp, which
B–adenovirus receptor (CAR). In the high-resolution is conserved within an otherwise variable region of protein

4 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Adenoviruses

IX VA RNAs E3
E1A E1B ML

L1 L2 L2 L4 L5
289R 55kDa gp19k10.4k14.5k
TP
A 243R 19kDa IX 52/55k IIIa III pVII V pµ pVI II Protease 100k 33k pVIII 11.6k 14.7k IV
A’
Ori Ori
IVa2 Pol pTP DBP ORF6 ORF4 ORF1
TP ORF6/7 ORF3 ORF2

E2 E4
IVa2

Figure 2 Organization of the human adenovirus 2 (Ad2) genome. The linear double-stranded DNA genome is represented by the pair of solid horizontal
lines in the centre of the figure. The terminal protein (TP) that is covalently linked to the 5’ end of each strand and the adjacent sequence required for
initiation of viral DNA synthesis (Ori) are indicated. The origins are included within an inverted terminal repeat sequence of 202 bp present at the ends
of the genome, designated A and A’. The locations of the eight RNA polymerase II transcription units are represented by barbed arrows drawn in the
direction of transcription, with the immediate early, early and late transcription (ML) units shown in pink, blue and red, respectively. Viral proteins encoded
within each transcription unit are listed above or below the genome, using the unsystematic nomenclature adopted in the field (see Tables 2 and 3). Several
virion proteins are synthesized as precursors that are proteolytically processed by the viral protease only following virion assembly. Such proteins are
indicated by the prefix p. The five families, L1 to L5, of ML proteins, which are defined by the locations of the 3’ ends of the mRNAs that specify them (see
Figure 5), are indicated. The coding sequences for proteins that perform related functions are often organized together in the viral genome. Thus, all viral
replication proteins, including the DNA polymerase (Po1), the protein primer and precursor to the TP (pTP) and the single-stranded DNA-binding
protein (DBP), are encoded in the E2 transcription unit, while coding sequence for the core proteins or their precursors, pVII, V and pm, lie within the L2
segment of the ML transcription unit. The positions of the virus-associated (VA) RNA genes transcribed by RNA polymerase III are indicated by the solid red
arrowheads.

III in many human serotypes. These Arg-Gly-Asp peripentonal hexons. It is also likely to contact the inner
sequences are located in five projections, one per subunit, nucleoprotein core.
that extend some 20 Å from the top surface of the penton In addition to the DNA genome, the inner cavity
base. Each penton base can bind five integrin molecules, contains the basic core proteins, V, VII and m, two copies of
and can therefore presumably induce clustering of the terminal protein (TP; Figure 2), and the viral protease
integrins in the plasma membrane to trigger internalization (Table 2 and Figure 1b). Multimers (of unknown stoichio-
of virus particles by endocytosis. metry) of protein VII are tightly associated with the viral
The major structural features of the hexon monomer DNA, while protein V occupies a more external location in
(Table 2), determined by X-ray crystallography of the Ad2 the core. The high arginine content and strong double-
hexon (Athappilly et al., 1994), are two copies of an eight- stranded DNA binding activity of protein m suggest that it
stranded b-sheet motif termed a b barrel. This motif is plays an important role in condensing viral DNA for
found in the major capsid proteins of a number of other packaging into virions. Although protein VII is the major
DNA and RNA viruses. In the hexon trimer, the b barrels DNA packaging protein, very little is known about how it
form a pseudohexagonal base that is well suited to the close binds to and organizes the viral genome. Indeed, no details
packing of each hexon with its six identical neighbours. of the structure of the core are presently available, for the
This hollow base supports three towers projecting outward viral nucleoprotein is not stable once released from virions,
from the surface of the capsid (Figure 1a). Extensive and appears disordered in images of virions reconstructed
intertwining of loops from each monomer in the tower from cryoelectron micrographs.
make the hexon a very stable structure.
The other four proteins of the capsid (IIIa, VI, IX and
VIII; Table 2) are believed to stabilize this structure. Such a
function is clearly established for protein IX, for particles Replication
that lack this protein are more thermolabile than their
wild-type counterparts. On the outer surface of the capsid, Most studies of adenovirus replication have employed
protein IX molecules lie along the entire length of the human tumour cells in culture synchronously infected by
interfaces among the hexons in the centre of each of the 20 Ad2 or Ad5, with approximately 104 virions produced per
faces of the icosahedral particle, as mortar between hexon cell within 2 days. Such efficient virus reproduction is
bricks (Figure 1b). Protein IIIa contacts hexons at the 30 achieved by redirection of the cellular systems responsible
edges of the icosahedron, where it extends from the exterior for synthesis and translation of mRNAs. The viral genome
to the interior of the capsid shell, like a protein rivet that contains coding sequences for at least 40 proteins, but only
fastens the edges between the faces of the capsid. Protein eight transcription units for cellular RNA polymerase II
VI, like protein VIII, is entirely internal and located at the (Figure 2). All but one (IX) of the viral transcription units is
vertices of the particle, where it anchors the ring of polycistronic, encoding from several to nearly 20 proteins.

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 5


Adenoviruses

This organization minimizes the viral genetic information 80–85% of the adenovirus particles that initially bind to a
devoted to transcriptional control signals, but requires cell enter endosomes. Their escape from these organelles
alternative processing of primary transcripts to produce into the cytoplasm via disruption of the endosomal
mRNAs for translation into the individual viral proteins. membrane is mediated by the penton base, by a mechanism
During the infectious cycle, viral proteins are made in a that has not been elucidated.
strict temporal sequence. By convention, early genes are During entry, the virus particle is sequentially disas-
defined as those expressed before viral DNA synthesis sembled, or uncoated. Within endosomes, the pentons and
begins. The early phase is devoted to synthesis of viral proteins IIIa and VIII first dissociate. Once particles
proteins needed for efficient expression of viral genes and lacking these proteins enter the cytoplasm, protein VI is
viral DNA synthesis, or that optimize the environment for degraded by the virion protease, a cysteine protease whose
virus reproduction. By contrast, structural proteins are activity appears to be regulated by redox potential. The
made only once the genome has replicated, during the late remaining stabilizing protein, protein IX, then dissociates,
phase of infection. allowing release of the core nucleoprotein into the
cytoplasm for transport to the nucleus, the site of viral
gene expression, replication and assembly. Adenoviral
Attachment, entry and uncoating nucleoproteins traverse nuclear pore complexes, presum-
The binding of the virion fibres to the integral membrane ably via the cellular nuclear protein import pathway.
protein CAR mediates the initial attachment of most Within the nucleus, the viral genome, which remains
human adenoviruses to susceptible cells. Detailed char- associated with protein VII, localizes to a limited number
acterization of fibre protein–CAR interactions has im- of specific sites, termed replication centres. These sites
plications for construction of peptide-based inhibitors of correspond to preexisting nuclear niches that are used by a
adenovirus infection (Kirby et al., 2000). The concentra- number of other viruses with DNA genomes.
tion and location of this receptor on cell surfaces is one
parameter that influences the tissue tropism of these viruses Early phase of infection
and their utility as recombinant vectors for therapy
(Shayakhmetov et al., 2000; Wickham, 2000). Initial The first biosynthetic reaction in the infectious cycle is
attachment via the fibre facilitates binding of penton bases synthesis of E1A pre-mRNA by RNA polymerase II under
to av-integrins, interactions that induce clustering of the direction of E1A control sequences that allow efficient
integrin-bound virions into coated pits and subsequent transcription in a variety of cell types. Alternative splicing
internalization by receptor-mediated endocytosis. Some produces mRNAs for two E1A proteins (Figure 3a).

13S Inactive
289R E2f + Rb Ad2 E2 early
Dp1
CR1 CR2 CR3 E2f site Cdk2
Cyclin E
12S Thymidine kinase
E1A Active
243R
CR1 CR2 E2f
Rb
p300
Rb E1A
(a) (b)

Figure 3 Structure and function of human adenovirus 2 (Ad2) E1A proteins. (a) Organization of E1A proteins. Alternative splicing of E1A pre-mRNA, in
which introns are indicated by the caret symbols, in the infected cell nucleus produces the abundant 13S and 12S E1A mRNAs. Because splicing
does not change the translational reading frame, the 289R and 243R E1A proteins translated from these mRNAs differ only in the internal sequence
of 46 amino acids unique to the 289R protein. This unique sequence contains most of one of the three highly conserved regions (CR1 to 3) identified by
comparison of E1A sequences of human adenoviruses. The regions of the E1A proteins necessary for binding to the cellular proteins Rb and p300 are
indicated. (b) Model for countermanding Rb protein-mediated regulation of transcription by E1A proteins. The E2f proteins are sequence-specific
transcriptional activators first identified by virtue of their binding to the adenoviral E2 early promoter. As indicated, these are heterodimeric proteins,
comprising one member of the E2f family, which contains six known members, and a Dp protein, such as Dp1. In actively growing mammalian cells, E2f
proteins are bound to hypophosphorylated Rb protein for most of the cell cycle, but free from mid-G1 through S phase, when Rb becomes heavily
phosphorylated. The Rb-E2f complexes retain DNA-binding activity and bind to E2f recognition sites in specific promoters. However, the Rb protein
actively represses transcription, as indicated by the red bar (top right). The adenoviral E1A proteins made in infected (or transformed) cells bind to the Rb
protein by means of a CR2 sequence and, via CR1 sequences, actively dismantle Rb-E2f complexes. Thus, the E1A protein-Rb interaction releases E2f
that can then stimulate transcription (green arrow, bottom right). This mechanism would make E2f available for transcription from the viral E2
promoter and from those of cellular genes normally expressed in the late G1 and S phases of the cell cycle. Such genes include those whose products
ensure progression through the cell cycle (e.g. cyclin-dependent kinase (Cdk) 2 and cyclins E and A), carry out replication of the cellular genome or produce
substrates for DNA synthesis, such as thymidine kinase and dihydrofolate reductase. Adenoviruses supply viral replication proteins, but depend on host cells
for the latter enzymes. The ability of E1A proteins to abrogate the negative regulatory function of the Rb protein is required for their transforming activity.
Adapted from Flint SJ et al. (2000) Principles of Virology: Molecular Biology, Pathogenesis and Control, ASM Press, Washington, DC, with permission.

6 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Adenoviruses

Mutations that impair production of the larger (289R) Viral DNA synthesis
E1A protein prevent efficient transcription of all other viral
genes. Thus, E1A expression defines an immediate early The adenoviral genome is replicated by continuous
phase of infection. The 289R E1A protein is the prototype synthesis of both new strands of the DNA from a protein
for viral transcriptional activators that stimulate RNA primer (Challberg et al., 1980) (Figure 4). A complex of the
polymerase II transcription, but do not bind specifically to viral DNA polymerase and preterminal protein (pTP)
DNA sequences in the template. It has therefore been binds specifically to the terminal origins of replication
studied extensively in simplified experimental systems, in (Figure 2) and the polymerase then catalyses covalent
which it stimulates transcription from an enormous variety linkage of deoxycytidine monophosphate (dCMP) to the
of viral, cellular and synthetic promoters. It can bind to pTP to provide the 3’-OH primer universally required for
several of the general initiation proteins required for template-directed DNA synthesis (Figure 4). The parental
specific transcription by RNA polymerase II and can strand initially displaced is replicated in the same way
operate by means of many cellular transcriptional reg- (Figure 4). The viral DNA-binding protein (DBP) (Table 3)
ulators that bind to specific promoter sequences. The 289R unwinds the double-stranded DNA template and acts as a
E1A protein may therefore function as a coactivator of processivity factor, allowing the DNA polymerase to move
broad specificity, facilitating interactions among cellular long distances along the template. Viral proteins carry out
transcriptional components and assembly of active RNA all the synthetic reactions needed for replication of the
polymerase II initiation complexes. adenoviral genome, but cellular transcriptional activators
Both E1A proteins also regulate transcription by (Figure 4), topoisomerases and enzymes that make the
binding to, and counteracting, the repression of transcrip- deoxyribonucleotide triphosphate substrates are also
tion of specific genes by the cellular retinoblastoma protein required. The rate of accumulation of viral replication
(Rb protein), the product of a tumour suppressor gene proteins and the availability of these cellular proteins are
(Figure 3b). This activity of E1A proteins is presumed to governed by the viral E1A proteins. Viral DNA synthesis
coordinate viral DNA synthesis with production of the begins 6–8 h after infection and continues exponentially as
necessary substrates. The E2f transcriptional activators to the products of one replication cycle serve as templates for
which Rb binds (Figure 3b) are required for efficient the next. The concentration of viral DNA molecules and
transcription of both the viral E2 transcription unit, which replication proteins at specific intranuclear sites must
encodes the viral replication proteins (Figure 2), and the facilitate efficient replication. Multiple factors may con-
cellular genes for enzymes that synthesize substrates for tribute to the cessation of viral DNA synthesis by about
DNA synthesis, upon which adenoviruses depend. Tran- 24 h after infection, including encapsidation of an increas-
scription of these cellular genes normally begins shortly ing fraction of newly synthesized DNA genomes as virion
before S phase, but in Ad2 infected cells would be induced proteins are made, exhaustion of the cellular substrate pool
in parallel with viral E2 transcription once E1A proteins and inhibition of synthesis of proteins needed for replica-
were available to remove Rb from Rb-E2f complexes tion late in the infectious cycle.
(Figure 3b).
Transcripts of the early E1B, E3 and E4 transcription Late phase of infection
units, like E2 pre-mRNAs, are processed by alternative
pathways to mRNAs specifying two or more proteins Viral DNA replication leads to synthesis of the structural
(Figure 2). These early proteins facilitate efficient viral proteins, most of which are encoded within the major late
replication in various ways (Table 3): some stimulate viral (ML) transcription unit (Figure 2). During the early phase
gene expression later in the infectious cycle; some block of infection, ML transcription ceases near the middle of the
E1A protein-induced apoptosis (programmed cell death); genome, rather than at the end of the transcription unit, the
and yet others protect the infected cell against immune pattern observed during the late phase. The mechanism
defences mounted by the host organism (Table 3). The early responsible for this unusual transcriptional change has not
phase of infection is also marked by production of a viral been established. The rate of ML transcription by RNA
countermeasure to a host defence that operates intracellu- polymerase II increases between 20- and 30-fold in the late
larly. The small virus-associated (VA) RNA I, one of two phase of infection. Such stimulation requires two infected
very abundant viral RNAs synthesized by cellular RNA cell-specific proteins that bind specifically to sequences
polymerase III (Figure 2), prevents inactivation of an within the first ML intron. One is a dimer of the viral IVa2
essential component of the translation initiation machin- gene product (Figure 2), while the other comprises the IVa2
ery (eIF2) by a general antiviral defence mechanism protein and at least one other, not yet identified protein.
(Table 3). Thus, VA RNA I is necessary for efficient Thus, synthesis of the IVa2 protein in the infected cell is
synthesis of viral late proteins (Thimmappaya et al., 1982). essential for efficient ML transcription. However, this
viral, sequence-specific transcriptional activator is itself the
product of a late gene (Figure 2). Temporal regulation of
IVa2 transcription appears to be effected by titration of a

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 7


Adenoviruses

Pol
5’
A
pTP Ser C OH A’
3’
3’ 5’
3’OH GpTpApGpT 5’
2 DBP
dNTPs
1 Pol
A
+ Nf1, Oct-1
TP pTP
A A’
5’ A’
5’ 5’
5’

Protease

A A’ A A’
5’ 5’
5’ 5’

5
3
Mutiple rounds
of replication
4 A
5’
5’
A’ A pTP- A’
Pol:
Nf1, Oct-1

Figure 4 Replication of adenoviral DNA. Assembly of a complex of the viral DNA polymerase (Po1) and pTP at the viral origins (step 1) at each end of the
genome, is facilitated by the cellular, sequence-specific transcriptional activators Nf1 and Oct-1, which bind to the origins and to the viral replication
proteins. Pol then catalyses covalent linkage of dCMP to a specific serine residue in pTP to provide the 3’-OH primer needed by all known DNA polymerases
(box, top left). This enzyme synthesizes viral DNA from this primer in the 5’!3’ direction (step 2), in a reaction that requires the viral DNA-binding protein
(DBP) (Table 3), which coats the displaced strand and unwinds the double-stranded template, and a cellular topoisomerase to relieve supercoiling of the
DNA ahead of the replication fork and torsional stress. As the genome carries an identical origin at each end, each parental strand can be replicated by this
continuous mechanism, with displacement of its complement. Such displaced strands carry the complementary, terminal sequences of the inverted
terminal repetition, designated A and A’. Their reannealing forms a short duplex stem identical to the terminus of the parental genome (step 3). Thus,
origins are reformed, allowing protein priming and continuous synthesis of the parental strands initially displaced (steps 4 and 5). From Flint SJ et al. (2000)
Principles of Virology: Molecular Biology, Pathogenesis and Control, ASM Press, Washington, DC, with permission.

cellular transcriptional repressor that binds to the IVa2 As viral late mRNAs accumulate, synthesis of cellular
promoter with exponential viral DNA synthesis. proteins is gradually but inexorably inhibited (Beltz and
Induction of the late pattern of transcription leads to Flint, 1979). Two posttranscriptional mechanisms con-
dramatic alterations in the processing of ML pre-mRNAs, tribute to such selective expression of viral genetic
some of the best understood of several examples of information. A complex of the E1B-55 kDa and E4
temporal regulation of viral RNA processing. Utilization ORF6 proteins (Table 3) induces selective export of viral
of both the five poly(A)-addition sites and of certain splice late mRNAs from the nucleus, by a mechanism that is not
sites in ML pre-mRNAs is altered following entry into the well understood. Viral late mRNAs are also preferentially
late phase of infection (Figure 5). All mRNAs processed translated. Hypophosphorylation of the cap-binding
from ML pre-mRNAs carry a common, 5’-terminal component of the translation initiation protein eIF4F
sequence formed by splicing of three small exons, the induced by a viral late protein, or proteins, inhibits the
tripartite leader sequence (TPL). As each ML transcript most common mechanism of initiation, scanning of
contains but a single copy of these exons, only one mRNA ribosomes from the 5’ cap of the mRNA to the initiation
can be made, with the sequences for all the others discarded codon. Translation of most cellular mRNAs is therefore
during processing. The presence of the TPL must therefore inhibited, but the TPL allows an alternative mechanism of
confer advantage(s) that outweigh the apparent ineffi- initiation, and thus the translation of ML mRNAs. The
ciency of this baroque mechanism of ML mRNA synthesis. viral L4 100-kDa protein, an RNA-binding protein, is

8 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Adenoviruses

Major late promoter


0 16.5 50 100 Map
units

Transcription
I1 I2 I3
Pre-mRNA 5’ C L1 L2 L3 L4 L5 3’

L1 L2 L3 L4 L5

Poly(A) addition at one of the L1 to L5 sites


C
C
C
C
C

Alternative splicing

C L1 An C L1 L2 L3 An
Two possible 3’ 4 possible 3’
splice sites splice sites

Tpl
C L1 An 52/55kDa C L3 An pIV
C L1 An IIIa C An Hexon
L1mRNAs C An Protease
C An
(a)
L3 mRNAs

L1 L2 L3
C Primary transcript
Cstf
3’ splice sites
C
SR

L1
C 52/55kDa mRNA

(b)

Figure 5 Alternative processing of major late (ML) pre-mRNA. The Ad2 genome is represented by the solid horizontal lines at the top of the figure, 0-100
map units, with the site of initiation of ML transcription by RNA polymerase II indicated by the jointed arrow drawn in the direction of transcription.
During the late phase of infection (a), ML transcription proceeds from this initiation site to close to the right-hand end of the genome. The large
(nearly 30 kb) primary transcript shown below the genome, with the 5’ cap designated (C), is processed into over 15 mRNAs by polyadenylation at one of
five possible sites, L1 to L5 (vertical arrows), used at approximately equal frequency. The polyadenylation sites define the L1 to L5 families of 3’ coterminal
mRNAs. The tripartite leader sequence present at the 5’ ends of all ML mRNAs is formed by splicing of three small exons, l1 to l3, and is then ligated to
alternative 3’ splice sites, as illustrated for the L1 and L3 mRNAs. During the early phase of infection (b), ML transcription terminates at multiple sites with a
large region around the middle of the transcription unit, so that no sequences beyond map unit 70 are transcribed. Even though these ML transcripts
contain the L1, L2 and L3 polyadenylation sites, the L1 site is preferentially utilized. As indicated, the splicing of L1 pre-mRNAs is also temporally regulated.
Only the 52/55-kDa mRNA is made during the early phase, but the 3’ splice site for the protein IIIa mRNA is also recognized during the late phase. Changes
in the activities of specific, cellular polyadenylation or splicing proteins induced by viral proteins appear to effect these alterations in ML pre-mRNA
processing. Adapted from Flint SJ et al. (2000) Principles of Virology: Molecular Biology, Pathogenesis and Control, ASM Press, Washington, DC, with
permission.

required for efficient initiation of translation of all viral late Assembly, maturation and release of virus
mRNAs. It remains to be determined how the actions of particles
this protein, the TPL and VA RNA I are integrated to
ensure efficient synthesis of viral proteins in a translation- Viral structural proteins enter the nucleus where hexons
ally compromised environment. and pentons assemble from their monomeric component
(Table 2) in reactions driven by the high concentrations of
the proteins. Formation of hexons trimers requires the L4

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 9


Adenoviruses

Table 3 Subgroup C adenovirus early products


Transcription unit Protein a or RNA Functions and properties
E1B 55kDa Promotes selective export of viral late mRNA from the nucleus
during the late phase as a complex with the E4 ORF6 protein;
binds to the cellular p53 protein to block apoptosis
19 kDa Blocks apoptosis
E2 DNA polymerase (Pol) Initiates viral DNA synthesis by catalysing covalent linkage of
dCMP to a serine residue in the pTP protein primer, when bound to
pTP at viral replication origins; completes continuous replication
of both strands of the genome
Preterminal protein (pTP) Protein primer for initiation of viral DNA synthesis; incorporated
into virions and processed to the TP by the virion L3 protease; TP
(and probably pTP) facilitate unwinding of the origin during
replication
Single-stranded DNA-binding Stimulates initiation of viral DNA synthesis and essential for
protein (DBP) elongation; binds single-stranded DNA cooperatively and with
high affinity probably as long protein chains, to promote
unwinding of the DNA template, stimulate activity of Pol up to
100-fold and induce highly processive DNA synthesis by the viral
enzyme; can stimulate transcription from viral early promoters;
required posttranscriptionally for efficient production of viral late
mRNAs in semipermissive simian cells
E3 gp19K Glycoprotein localized to membrane of the endoplasmic reticulum
(ER) where it binds MHC class I proteins and prevents their
transport to the cell surface; can therefore block cell killing by
cytotoxic T cells
RID (receptor internalization Heterodimeric integral membrane protein localized to plasma
and degradation) (10.4 K and membrane, Golgi and ER; induces endocytic internalization and
14.5K) degradation of cellular ‘death domain’ containing receptors that
signal apoptosis when activated, to block this response; can
stimulate internalization of receptor protein tyrosine kinases
ADP (adenovirus death Glycoprotein localized to the nuclear membrane, produced in large
protein) (11.6K) quantities late in infection; required for efficient release of progeny
virions from the nucleus and lysis of the infected cell
12.5K Not known
6.7K Not known
E4 ORF1 Not known
ORF2 Not known
ORF3 Increases stability of unprocessed major late pre-mRNA in the
nucleus; required for proper splicing of these pre-mRNAs; can
promote exon inclusion during splicing; required for maximally
efficient viral DNA synthesis, especially when the E4 ORF6
protein cannot be made; in the absence of both this and the ORF6
protein, abnormal concatameric viral DNA molecules are
produced; induces reorganization of nuclear structures containing
cellular transcription and replication proteins
ORF4 Binds to cellular protein phosphatase 2A (PP2A) to induce
hypophosphorylation of E1A proteins and specific cellular
proteins; negatively regulates E1A and E4 transcription in infected
cells via PP2A
continued

10 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Adenoviruses

Table 3 – continued
Transcription unit Protein a or RNA Functions and properties
E4 ORF6 Like the ORF3 protein, increases nuclear stability of major late
pre-mRNAs and is required for qualitatively and quantitatively
normal viral DNA synthesis; can promote exon exclusion during
pre-mRNA splicing; in complex with the E1B 55-kDa protein,
promotes selective export of viral late mRNAs from the nucleus
during the late phase of infection; contains a nuclear export signal
and can shuttle between the nucleus and the cytoplasm; binds to
the cellular p53 protein and stimulates its degradation
ORF6/7 Can stimulate cooperative binding of cellular E2f transcriptional
activators to a pair of inverted binding sites in the early E2
promoter
ML L1 52/55 kDa Required for assembly of virions, encapsidation of the DNA
genome
VA RNA I VA RNA I RNA of 166 bp synthesized by RNA polymerase III that
accumulates to ~108 copies per cell; binds to the cellular
double-stranded RNA-dependent protein kinase (PKR); prevents
activation of PKR and hence phosphorylation and inactivation of
eIF2, which is essential for initiation of translation
ML, major late; VA, virus-associated.
a
Viral early proteins are listed by the unsystematic names in common use. These names are derived from the protein’s properties or functions
(E2, E3), apparent molecular mass based on electrophoresis in SDS-polyacrylamide gels (E1B, E3, L1) or the position of coding sequences
within the transcription unit (E4).
Refer to Figure 5 for explanation of L1 early products.

100-kDa protein. Virus particles then assemble by either a Epidemiology


sequential or a concerted mechanism (Figure 6). The viral
L1 52/55-kDa, the L4 33-kDa and the IVa2 proteins assist Because adenovirus infections in humans are not easily
the complex process of assembly in ways not yet well identified by purely clinical criteria, the epidemiology of
understood. The encapsidation of viral DNA is directed by these viruses has been studied using serological or
packaging signals located near the left end of the genome molecular methods of virus detection. Our current under-
(Figure 6). The initially assembled immature particles standing is based on studies in various selected populations
contain precursors to six virion proteins and are not and worldwide data on isolation of adenoviruses, collected
infectious. Their maturation to infectious particles requires by the World Health Organization.
the L3 protease, which is incorporated into assembling Adenoviruses are ubiquitous pathogens that infect
particles and cleaves these precursors at specific sites after humans of all ages and races, with few gender-specific
assembly. differences. They are transmitted by direct contact, the
Adenovirus infection inhibits production of cellular faecal–oral route and, in some cases, in water. Members of
macromolecules as cellular systems and resources are subgroup C, and probably subgroup F, are endemic in all
redirected for viral replication. Consequently, the infected populations examined, with 80–100% of children infected
cell eventually dies, with release of progeny virions. Specific within the first 3 years of life. By contrast, other human
viral proteins may actively promote such cell destruction or adenoviruses are associated with epidemics, for example,
lysis. The E3 adenovirus death protein (ADP) can disrupt of acute respiratory disease (ARD) among military recruits
the nuclear membrane (Table 3) and the viral protease (Table 1). Parameters that influence transmission and
cleaves certain proteins of the cytoskeleton. The inability incidence of infection include population density, crowd-
of the infected cell to maintain its architectural integrity ing and stress, socioeconomic status and the time of year.
once cellular protein synthesis has been inhibited may Individuals with impaired cell-mediated immunity, such as
account for the dependence of release of virions on such transplant patients receiving immunosuppressive drugs,
inhibition. are at higher risk for severe adenovirus infection.

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 11


Adenoviruses

Cytoplasm Nucleus L1 52/55-kDa protein


Empty
capsid Assembly
intermediate
L4
100-kDa
protein

Protein II

Hexon trimer

Other virion proteins Newly synthesized pIIIa


pTP
viral DNA pVI
pVII
Young pVIII
virion pµ
B

L3 protease

Protein
III
Protein IIIa
IV TP
Penton VI
VII
Virion VIII
µ

Figure 6 Assembly of adenovirus particles. Virion proteins (Table 2) are synthesized in large quantities in the cytoplasm of infected cells and imported into
the nucleus. The major structural units of the virion, hexons and pentons, then assemble from their monomeric components. Pentons are formed by self-
assembly, but hexons can be assembled only with the assistance of the viral L4 100-kDa protein, which binds to hexon monomers. Two possible pathways
for capsid assembly are depicted. In the sequential assembly pathway (A), hexons, pentons and capsid-stabilizing proteins self-assemble into empty
capsids, which contain the L1 52/55-kDa proteins. These L1 proteins may form a scaffold for capsid assembly, and are required for encapsidation of the
genome. In this mechanism, newly-synthesized viral DNA is inserted into preformed empty capsids upon recognition of a packaging sequence that lies at
the left end of the genome. Core proteins enter the capsid with viral DNA to form noninfectious, immature particles (young virions). Cleavage of the
precursors to the six virion proteins listed at the right by the L3 protease, produces infectious virions. The failure of an Ad5 mutant with a deletion within the
packaging sequence to direct assembly of any capsid-like structures indicates that assembly of the capsid and encapsidation of the genome may be
concerted reactions (pathway B). The incomplete particles shown in pathway A would then represent ‘dead-end’ products that cannot complete assembly,
or artefacts of the methods of extraction of particles from infected cells. Adapted from Flint SJ et al. (2000) Principles of Virology: Molecular Biology,
Pathogenesis and Control, ASM Press, Washington, DC, with permission.

Clinical Features children, especially those who are malnourished or


suffering from measles. Statistics on death resulting from
Adenoviruses rarely cause fatal disease, and as many as viral disease identify Ad7 as the most pathogenic of the
30–50% of infections may be asymptomatic. Nevertheless, human adenoviruses.
these viruses are important causes of respiratory disease, The first syndrome to be associated with adenoviruses
conjunctivitis and gastroenteritis (Table 1). The subgroup C was ARD in military recruits. Ad4 (subgroup E), and less
serotypes commonly cause acute upper respiratory disease frequently Ad7 and other members of subgroup B, are the
in young children, and they and subgroup B members primary causes, with rates of morbidity as high as 6–17 per
account for 2–7% of lower respiratory illness in this 100 per week among recruits in North America and
population, with such symptoms as bronchitis, pharyngitis Europe. Other well-characterized epidemic diseases caused
and pneumonia. The latter syndrome, which may be fatal, by specific adenoviruses are pharyngoconjunctival fever
is most usually associated with Ad7 infection of very young (PCF) and epidemic keratoconjunctivitis (EKC) (Table 1),

12 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net


Adenoviruses

in which the virus is spread by contact with contaminated for this activity (Whyte et al., 1988) (Figure 3b). The E1A
water, such as in contaminated ponds and inadequately protein-induced release of E2f transcriptional activators
chlorinated swimming pools, or with contaminated from association with Rb (or related) protein is believed to
instruments (or hands) in medical and ophthamological be primarily responsible for the mitogenic activity of these
facilities, respectively. The former disease is associated viral proteins, and the uncontrolled proliferation of E1A-
with both respiratory tract illness and conjunctivitis. In transformed cells. Unscheduled DNA synthesis induced in
contrast, EKC is characterized by conjunctivitis, followed this way activates control mechanisms that inhibit cell
by corneal infiltration, which may impair vision for several cycle progression or activate apoptosis via the product of
months. the cellular p53 gene, which is the most frequently mutated
Several adenoviruses can cause diarrhoea (Table 1). The gene in human tumours. When potentially genotoxic
subgroup F viruses are important causes, second only to damage is detected, the normally low concentration of
rotaviruses, of acute gastroenteritis in young children p53 increases substantially and, by stimulation of tran-
throughout the world. Infected children usually exhibit scription of specific genes, p53 induces arrest of the cell
clinically moderate disease with diarrhoea, vomiting and cycle in G1 or apoptosis. The E1A proteins induce
dehydration, but the diarrhoea may last for 1–2 weeks and accumulation of the p53 protein by at least two mechan-
fatal infections by Ad40 or Ad41 have been recorded. isms, and apoptosis. However, both proteins encoded
Much less commonly, adenovirus infection leads to within the viral E1B transcription unit can block this
severe complications, such as encephalomeningitis, intus- response. Thus, the E1B proteins are necessary for
susception (the prolapse of one section of the intestine into complete transformation because they prevent cells
the neighbouring portion; predominantly with Ad13 and synthesizing E1A proteins from committing suicide.
Ad5) in infants, and fatal neonatal infections with systemic This model accounts for the activities exhibited by the
symptoms and pneumonia. viral E1A and E1B transcription units in transformation
assays, and is based on a large body of biochemical,
molecular and genetic data. Nevertheless, other mechan-
isms may contribute to adenovirus transformation: E1A
Viral Oncogenesis sequences encoded within the second exon (Figure 3a) are
required for immortalization of primary cells in culture,
Human adenovirus 12 (Ad12) and Human adenovirus 18 and E1A proteins can bind to other cellular proteins that
(Ad18) were the first human viruses shown to be oncogenic negatively regulate cell cycle progression.
(Trentin et al., 1962). There is no evidence that any
adenovirus contributes to tumour development in humans,
but the molecular mechanisms by which viral proteins alter Differential oncogenicity of human
the function of host cell tumour suppressor proteins are adenoviruses for rodents
similar to those of human papillomaviruses causally
associated with cervical cancer in women. The striking Adenoviruses that are highly oncogenic or nononcogenic
differences in the oncogenicity of subgroup A and C in rodents (Table 1) do not differ markedly in the
adenoviruses (Table 1) has allowed investigation of frequencies with which they transform cultured rodent
mechanisms that determine cell growth and proliferation cells, nor can cells transformed by them be distinguished by
in vivo. Furthermore, most human adenoviruses transform their in vitro properties. However, rat cells transformed by
rodent cells in culture. the subgroup A Ad12 induce tumours in newborn, and
sometimes adult, syngeneic rats, whereas those trans-
Mechanisms of transformation formed by the subgroup C Ad2 or Ad5 do not, and are
tumorigenic only in experimentally immunosuppressed
The products of the adenoviral E1A and E1B transcription rats or immunodeficient nude mice. Such observations
units transform rodent cells. For example, these viral DNA indicate that the host’s immune response mediated by
sequences can induce full transformation when introduced cytotoxic T cells (CTLs) eliminates subgroup C adeno-
into cells in the absence of additional viral genetic virus-transformed cells much more effectively than those
information. In such DNA-mediated transformation transformed by subgroup A members. Indeed, Ad5 and
assays, E1A DNA is necessary, and in some cases, Ad12 transformants derived from the same parental rat
sufficient. Complete transformation generally requires cells are susceptible and resistant, respectively, to lysis by
the adenoviral E1B transcription unit; however, E1B appropriate CTLs in vitro. This difference is determined by
sequences alone exhibit no transformation activity what- the viral E1A proteins. Ad12 E1A proteins may confer
soever. resistance to killing by CTLs by reducing cell surface
Our current understanding of the mechanism of E1A concentrations of major histocompatibility complex
protein-dependent transformation is based on identifica- (MHC) class I proteins, which present proteasome-
tion of cellular proteins that bind to E1A regions required processed viral antigens (peptides) on the exterior of the

ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net 13


Adenoviruses

cell for recognition and subsequent cell killing by CTLs. Rowe WP, Huebner RJ, Gilmore LK, Parrott RH and Ward TG (1953)
The Ad12, but not Ad5, E1A proteins repress transcription Isolation of a cytopathogenic agent from human adenoids undergoing
of MHC class I genes. The nature of the E1A protein spontaneous degeneration in tissue culture. Proceedings of the Society
for Experimental Biology and Medicine 84: 570–573.
epitopes presented to the immune system may also
Shayakhmetov DM, Papayannopoulou T, Stamatoyannopoulos G and
influence CTL recognition. Lieber A (2000) Efficient gene transfer into human CD34+ cells by a
Other components of the immune system have been retargeted adenovirus vector. Journal of Virology 74: 2567–2583.
implicated in the rejection of cells transformed by Thimmappaya B, Weinberger C, Schneider RJ and Shenk T (1982)
subgroup C adenoviruses. The degree of resistance of Adenovirus VAI RNA is required for efficient translation of viral
adenovirus-transformed cells to natural killer (NK) cells, mRNAs at late times after infection. Cell 31: 543–551.
important components of the innate, ‘nonspecific’ immune Trentin JJ, Yabe Y and Taylor G (1962) The quest for human cancer
viruses. Science 137: 835–849.
response, also correlates with tumorigenicity. This differ-
Whyte P, Buchkovich KJ, Horowitz JM et al. (1988) Association
ential response is also conferred by viral E1A proteins. between an oncogene and an anti-oncogene: the adenovirus E1A
Specific segments of subgroup C Ad E1A proteins render proteins bind to the retinoblastoma gene product. Nature 334: 124–
transformed cells susceptible to apoptosis induced by NK 129.
cells. Although the mechanism is not fully understood, Wickham TJ (2000) Targetting adenovirus. Gene Therapy 7: 110–114.
binding of the E1A proteins to p300/Cbp (Figure 3a)
appears to be important. A segment of the Ad12 E1A
proteins that is absent from E1A sequences of members of
subgroup C, and which does not modulate resistance of Further Reading
Ad12-transformed cells to either CTLs or NK cells, has
also been identified as an important determinant of Baum SG (2000) Adenovirus. In: Mandell GL, Bennett JE and Dolin R
tumorigenicity. Finally, although the tumorigenicity of (eds) Mandell, Douglas and Bennett’s Principles and Practice of
Infectious Diseases, 5th edn, pp. 1624–1630. Philadelphia: Churchill
transformed cells in immunocompetent syngeneic rats is Livingstone.
determined primarily by the viral origin of the E1A Burnett RM (1997) The structure of adenovirus. In: Chiu W, Burnett
transcription unit, cells that synthesize Ad12 E1B proteins RM and Garcea RL (eds) Structural Biology of Viruses, pp. 209–238.
are significantly more tumorigenic than matched cells New York: Oxford University Press.
making Ad5 E1B proteins. The complexities of the D’Halluin JC (1995) Virus assembly. Current Topics in Microbiology and
interactions among host defence systems and adenovirus- Immunology 199 (Pt 1): 47–66.
transformed cells therefore remain far from completely Flint J and Shenk T (1989) Adenovirus E1A protein paradigm: viral
transactivator. Annual Review of Genetics 23: 141–161.
understood. Greber UF (1998) Virus assembly and disassembly: the adenovirus
cysteine protease as a trigger factor. Reviews in Medical Virology 8:
References 213–222.
Morsey MA and Caskey CT (1999) Expanded capacity adenoviral
Athappilly FK, Murali R, Rux JJ, Cai Z and Burnett RM (1994) The vectors: the helper-dependent vectors. Molecular Medicine Today 5:
refined crystal structure of hexon, the major coat protein of adenovirus 18–24.
type 2, at 2.9 Å resolution. Journal of Molecular Biology 242: 430–455. Nemerow GR and Stewart PL (1999) Role of alpha(v) integrins in
Bailey A and Mautner V (1994) Phylogenetic relationships among adenovirus cell entry and gene delivery. Microbiology and Molecular
adenovirus serotypes. Virology 205: 438–452. Biology Reviews 63: 725–734.
Beltz GA and Flint SJ (1979) Inhibition of HeLa cell protein synthesis Nevins JR (1992) E2F: a link between the Rb tumor suppressor protein
during adenovirus infection. Journal of Molecular Biology 131: 353– and viral oncoproteins. Science 258: 424–429.
373. Ruuskanen O, Meurman O and Akusjärvi G (1997) Adenoviruses. In:
Berget SM, Moore C and Sharp PA (1977) Spliced segments at the 5’ Richman DD, Whitley RJ and Hayden FG (eds) Clinical Virology, pp.
terminus of adenovirus 2 late mRNA. Proceedings of the National 525–547. Philadelphia: Churchill Livingstone.
Academy of Sciences of the USA 74: 3171–3175. Schneider RJ (1995) Cap-independent translation in adenovirus infected
Challberg MD, Desiderio SV and Kelly TJ (1980) Adenovirus DNA cells. Current Topics in Microbiology and Immunology 203: 117–129.
replication in vitro: characterization of a protein covalently linked to Shenk TE (1996) Adenoviridae: the viruses and their replication. In:
nascent DNA strands. Proceedings of the National Academy of Fields BN, Howley PM, Knipe DM et al. (eds) Fundamental Virology,
Sciences of the USA 77: 5105–5109. 3rd edn, pp. 979–1016. New York: Lippincott-Raven.
Chow LT, Gelinas RE, Broker TR and Roberts RJ (1977) An amazing Van der Vliet PC (1995) Adenovirus DNA replication. Current Topics in
sequence arrangement at the 5’ ends of adenovirus 2 messenger RNA. Microbiology and Immunology 199 (Pt 2): 1–30.
Cell 12: 1–8. Williams J, Williams M, Liu C and Telling G (1995) Assessing the role of
Kirby I, Davison E, Beavil AJ et al. (2000) Identification of contact E1A in the differential oncogenicity of group A and group C human
residues and definition of the CAR-binding site of adenovirus type 5 adenoviruses. Current Topics in Microbiology and Immunology 199 (Pt
fiber protein. Journal of Virology 74: 2804–2813. 3): 149–175.

14 ENCYCLOPEDIA OF LIFE SCIENCES / & 2001 Nature Publishing Group / www.els.net

You might also like