Vous êtes sur la page 1sur 37

Critical Reviews in Microbiology, 28(3):149–185 (2002)

Immune Response to Poxvirus Infections in


Various Animals
Scott A. Smith and Girish J. Kotwal*
Department of Microbiology and Immunology, University of Louisville School of
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

Medicine, Louisville, KY

* Address correspondence to: Girish J. Kotwal, Division of Medical Virology, Univ. of Capetown Medical
School, Cape Town, South Africa 7925. e-mail GJOTW01@yahoo.com

ABSTRACT: The study of infections of vertebrate animals by poxviruses has remained a dynamic
area of research for the last century. The host range of poxviruses vary from extremely narrow to
exceedingly broad, and they have been shown to enter their host by either the respiratory route or
through the skin. The severity of infection varies dramatically from one species to another, causing
anywhere from a local, self-limiting infection, to a devastating systemic disease, such as smallpox.
Although the immune response to poxvirus infections are very similar to that seen in other viral
infections, the poxviruses, unlike most other viruses (with the exception of Herpes viruses), are able
to defend themselves. They have been shown to carry a repertoire of proteins involved in immune
evasion and immune modulation. Poxviruses encode proteins involved in blocking many of the
For personal use only.

strategies employed by the host to combat viral infections; they encode for proteins that block
activity of many chemokines, cytokines, serine proteases, and even complement. Traditionally,
different animal models have been used to study the pathogenesis of poxvirus infections, and the
characterization of virulence genes using mutant poxviruses. Additionally, new animal models are
being developed to study the possible therapeutic uses many of these poxvirus immune modulating
proteins might have. This review discusses the host immune response against poxvirus infections
in various animals, the viral counter response to the host, and the animal models used to study
poxvirus infection and immune modulating proteins.

GLOSSARY:

ADCC antibody-dependent cell-mediated cytotoxicity


C4b-BP complement 4b binding protein
CAM chorioallantoic membrane
CCR CC-chemokine receptor
CPV cowpox virus
CR complement receptor
CrmA cowpox cytokine response modifier A
CTL cytotoxic T-lymphocyte
CXCR CXC-chemokine receptor
CX3CR CX3C-chemokine receptor
DAF decay-accelerating factor

1040-841X/02/$.50
© 2002 by CRC Press LLC

149
EV ectromelia virus
fH factor H
FITC fluorescein-isothiocyanate
FPV fowlpox virus
GIF granulocyte-macrophage colony-stimulating factor-inhibitory factor
GM-CSF granulocyte-macrophage colony-stimulating factor
HIV human immunodeficiency virus
HS heparan sulfate
ICE interleukin-1β-converting enzyme
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

IFN interferon
IFNR interferon receptor
IL interleukin
IL-1R interleukin 1 receptor
IL-18BP interleukin 18 binding protein
IMP inflammation modulatory protein
MAC membrane attack complex
MCCH molluscum contagiosum chemokine homolog
MCP membrane cofactor protein
MCV molluscum contagiosum virus
MGF myxoma virus growth factor
For personal use only.

MHC major histocompatibility complex


MIP macrophage inflammatory protein
MV myxoma virus
MVA modified vaccinia Ankara
NF nuclear factor
NK natural killer
ORF open reading frame
PAECs pig aortic endothelial cells
RCA regulators of complement activation
SCR short consensus repeat
serpin serine protease inhibitor
SFV Shope fibroma virus
sTNFR soluble tumor necrosis factor receptor
TGF transforming growth factor
TNF tumor necrosis factor
TNFR tumor necrosis factor receptor
tPA tissue-type plasminogen activator
uPA urokinase-type plasminogen activator
VCP vaccinia virus complement control protein
vIFNR viral interferon receptor
vIL viral interleukin

150
vIL-1R viral interleukin 1 receptor
vIL-18BP viral interleukin 18 binding protein
vMIP viral macrophage inflammatory protein
vTNFR viral tumor necrosis factor receptor
VV vaccinia virus
VGF vaccinia virus growth factor
XCR C-chemokine receptor
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

I. INTRODUCTION cuss the Chordopoxvirinae.3 There are eight


genera of vertebrate poxviruses (see Table 1).
The poxvirus family, Poxviridae, has the Poxviruses have been found to infect verte-
largest virions of any animal virus family. brates ranging from birds to humans. Some of
They appear oval or brick shaped, approxi- these infections can be highly fatal; smallpox
mately 350 nm in length by 270 nm wide. virus (variola), which replicates only in hu-
For this reason, vaccinia was the first animal mans, has killed more people than any other
virus to ever be seen microscopically in 1886. infectious disease. In addition, myxoma vi-
Poxviruses contain a large linear double- rus, which infects rabbits, can have as high as
stranded DNA genome of 130 to 280 kbp. a 99% mortality rate. Much of the success of
This large genome is responsible for encod- poxviruses as pathogens is because of the
ing all the genes necessary for their unique immune evasion strategies they employ.1
cytoplasmic lifestyle. The members of the The poxvirus infection generally follows
For personal use only.

poxvirus family replicate almost entirely in- one of two courses, a localized infection
dependent of the infected cell nucleus. There- resulting in benign skin lesions, or a sys-
fore, being unable to utilize the host cell’s temic infection, resulting in viral dissemina-
enzymes to synthesize RNA or replicate tion and usually death. Local pathogenesis
DNA, they must encode for these important of molluscum contagiosum virus (MCV) and
proteins in their own genome. In addition to Shope fibroma virus (SFV) infection has been
those proteins encoded by poxviruses re- well documented. Although the pathogen-
quired for transcription, replication, and esis and host response to infection is quite
virion assembly, there are several nonessen- different, both poxvirus diseases are self-
tial proteins employed to establish a suitable limiting and remain localized to the skin.
habitat. Several interesting nonessential pro- The generalized infections, however, are
teins encoded by poxviruses include: comple- characterized by several stages, ending in an
ment control protein (VCP), interleukin one explosion of viral lesions. Ectromelia virus
receptor homolog, tumor necrosis factor re- infection of mice starts with an infection of
ceptor homolog, and interferon receptor ho- the skin, usually through the footpad. After
molog.1 These nonessential proteins, and some replication, the virus spreads through
several others, play an important role in the local lymphatics to the bloodstream, re-
modulating the host’s immune response to sulting in primary viremia. The virus then
the infection, thereby maintaining the viral replicates to high titers in the spleen and
habitat.2 liver before reentering the blood, resulting
The family Poxviridae is divided into two in a secondary viremia. Finally, the virus
subfamilies, Chordopoxvirinae, which are spreads to the skin, and a severe rash of
vertebrate poxviruses, and Entomopoxvirinae, ulcerating lesions very rapidly overcomes
which are insect poxviruses — we only dis- the mouse. The disease progression of vari-

151
TABLE 1
Poxvirus Classification. The Family Poxviridae, Subfamily Chordopoxvirinae, is
Composed of Eight Genera. Listed Are the Most Studied Members of Each Genus
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

ola virus infection in humans occurs simi- the infection processes and genes involved
larly, with high mortality rates. in interactions with the host, several animal
The immune response to poxvirus infec- models have been developed to study vari-
tion is very complex. Each arm of the im- ous poxvirus diseases and recombinant mu-
mune system plays a vital role in the segre- tant poxviruses. Animal infection models are
For personal use only.

gation and the elimination of a poxvirus important for testing the efficacies of new
infection. Early in the infection, the innate antiviral drugs or to explore new treatment
immune response attempts to keep the infec- strategies. Models have been developed to
tion under control, while the acquired im- study the inflammatory response generated
mune response is mounted. Complement, by poxvirus infection, analyzing the degree
interferon, NK, and inflammatory cells help of inflammation and cellular infiltration both
control the early infection. Within a few days, quantitatively and qualitatively. This allows
a very strong humoral immune response is for a better understanding of the genes asso-
mounted, and poxvirus-specific antibodies ciated with poxvirus pathogenesis and re-
are produced. The antibodies can then help sults in better characterization of mutant
control the infection through a variety of strains. Understanding pathogenesis assists
mechanisms, including virus neutralization in the generation of more attenuated vaccine
and complement activation, obsonization, and strains and poxvirus vectors for use in live
ADCC. In addition, a cell-mediated immune vaccines in hopes of reducing complications
response is mounted a few days into the associated with traditional vaccines. Animal
infection. Poxvirus-specific CTLs are gen- models have also been instrumental in the
erated and attack virus infected cells, ulti- study of poxvirus immune modulating pro-
mately resulting in clearance of the poxvirus teins. Several poxvirus immunomodulatory
infection. proteins have been investigated extensively
Animal models have been used quite for their potential use as therapeutic agents.4
extensively for the study of poxvirus disease These immune blocking proteins have very
pathogenesis and exploring the therapeutic powerful antiinflammatory properties that
potential of poxvirus immunomodulatory may be employed for treatment of many
proteins. To gain a better understanding of injuries or diseases that involve excessive

152
inflammation. Animal model systems have species whether a poxvirus remains localized
been used to test the value of these proteins or disseminates. Myxoma virus, for example,
for use in transplantation, vascular injury, produces benign fibromas in wild rabbits, but
CNS injury, sepsis, arthritis, and restenosis. spreads explosively in domestic rabbits, result-
The therapeutic effects of these proteins, ing in a very high mortality. Several portals of
revealed by these model systems, are often entry into the host have also been described
quite fascinating. previously for the different poxvirus infections.5
Entry into the host via the skin has been re-
ported for myxoma virus and SFV. The respi-
II. POXVIRUS INFECTIONS ratory route, however, is best documented for
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

ectromelia virus, vaccinia virus, and variola


Poxviruses have been found to cause an virus. Of the huge number of poxvirus infec-
acute infection in a large variety of animal tions, only a few were chosen to represent the
species. The host range and lethality of the various infection properties of the poxvirus
poxvirus infection varies significantly. Some family. Here we describe briefly the pathogen-
poxviruses can infect a wide range of animals esis and/or special features of several well-
(e.g., vaccinia), whereas others have a very studied poxvirus infections.
limited host range (e.g., variola) (see Figure 1).
Depending on the poxvirus and the host, some
infections remain local (e.g., MCV), while oth- A. Ectromelia Virus Infection of Mice
ers are disseminated throughout the organism
resulting in higher fatality rates (e.g., ectrome- Ectromelia virus (EV) infection and patho-
For personal use only.

lia). In some cases, it depends on the host genesis has been studied extensively in mice.6

FIGURE 1. Pathology in various animals. (A) Characteristic smallpox (variola major) rash on the face and hand
(B) of a young boy several days into the illness, before lesions ulcerate. (World Health Organization, with
permission.) (C) Dome-shaped MCV papules on chest and around areola.127 (D) Human CPV infection.
Lesions are very painful and appear brownish with central vesicles.26 (E) Pig infected intravenously with SPV
14 days post-infection, and showing generalized ulcerating rash.33

153
After infection through lesions of the footpad terial infections. In the 1950s, MV was de-
with a virulent strain, the disease pathogen- liberately released in southern Australia to
esis follows a well-documented disseminated control overpopulations of feral European
course. After the virus replicates rapidly at rabbits. Initially it was a success, killing
the site of entry, it will pass through the lym- masses of pest rabbits, but soon resistance
phatic system to the blood. The primary vire- grew and the rabbits repopulated.
mia then results in infection of the liver and
spleen, often within 5 days of the infection.
The virus then disseminates throughout the C. Shope Fibroma Virus Infection of
mouse, resulting in a secondary viremia, and Rabbits
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

will grow to high titers in the epidermis and


hair follicles, resulting in the characteristic Shope fibroma virus (SFV), a close rela-
rash. The mouse is most infectious between 6 tive of myxoma, has a natural host range that
and 21 days postinfection. In some mice, is limited only to eastern North American
BALB/c, for example, the infection is very cottontail rabbits. Transmitted by arthropod
severe and death will usually result 6 to 7 vectors, SFV infection develops into a local-
days after infection because of sever liver ized (at the site of inoculation) benign tu-
necrosis. Death occurs so quickly in these mor; the tumorigenic effect it causes on its
mice that the rash does not even have time to host varies depending on the virus strain.9
develop. In other strains of mice, most nota- Some strains exhibit disseminating effects
bly C57BL, the mortality is not as high.7 In and can be found in the spleen, liver, and
this case, the virus does not grow to as high a kidneys, while others remain noninvasive
For personal use only.

titer and disseminates more slowly. The mouse and show no signs of even locally spreading.
will often survive with no appearance of a Independent of the infecting strain, the virus
rash. is cleared by the immune system after ap-
proximately 2 weeks, and the rabbit recov-
ers.
B. Myxoma Virus Infection of
Rabbits
D. Vaccinia Virus Infection
Myxoma virus (MV) causes a very le-
thal disease, myxomatosis, in the European Vaccinia virus (VV) is the most studied
rabbit. Mosquitoes and fleas normally trans- of the poxvirus family. Although its original
mit MV passively, causing a persistent be- (natural) host remains to be found, it has a
nign, nondisseminated disease in its natural broad host range, causing mild or occasion-
host, the North and South American rabbit. ally severe infection in a large number of
However, when passed onto the European animals.10 Several feasible theories exist as
rabbits, it causes a lethality of over 99%. to the origin of vaccinia virus. VV is be-
After infection of a European rabbit, the MV lieved to have originated either from evolu-
replicates rapidly at the infection site and tion of variola virus, evolution of cowpox
then spreads systemically, resulting in infec- virus, or possibly a hybrid of variola and
tion of many tissues and development of cowpox viruses.5 Regardless of its origin,
myxomas (secondary lesions).8 Extensive vaccinia virus has played a central role in the
lesions form throughout the areas of infec- eradication of smallpox, conferring long-term
tion, both internally and externally, often immunity to variola virus infection by cross-
giving rise to secondary Gram-negative bac- reacting antibodies and cytotoxic T-lympho-

154
cytes. VV is also employed for the expres- cated, the last reported case occurring in
sion of foreign genes used to vaccinate Somalia in 1977.
against other disease-causing agents, such as
rabies. Although most human infections, or
immunizations, result in a very mild, self- F. Orf Virus Infection of Sheep,
limiting local infection, occasionally dissemi- Goats, and Humans
nation occurred, and the development of a
very serious and sometimes fatal neurologi- Orf virus infects the epithelium, forming
cal side effect ensues, known as post-vac- acute pustular lesions, primarily of sheep
cinial encephalitis.11 In most cases, however, and goats. Occasionally orf is zoonotic and
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

VV infections remain localized to the infec- is transmitted to humans, often those that
tion site and usually ends in complete recov- frequently handle sheep. Human infections
ery. The highly attenuated vaccinia strain, are often on the hand or finger and rarely
modified vaccinia Ankara (MVA), is cur- require medical treatment. Antibiotics are
rently a potential vaccine candidate. The often given to prevent a possible secondary
sequence of MVA revealed 25 split and/or bacterial infection from arising. Primary in-
mutated genes that result in truncated pro- fection in sheep is characterized by vesicle
teins, and thereby diminished virulence.12 formation, followed by pustulation, scabbing,
and finally resolution after about 4 weeks.
Although the immune response to this local
E. Variola Virus Infection of Humans infection is reportedly vigorous, both hu-
moral and cell-mediated, reinfection often
For personal use only.

Variola virus is the etiological agent for occurs.13 Secondary infections, however, are
the devastating disease smallpox. Two strains often resolved more rapidly, in 1 week, with
of variola virus exist, variola major and vari- vesicle and scab formation occurring more
ola minor. Variola major was associated with quickly.14 Recently, CD4+ T-lymphocytes
high fatality rates of 5 to 40%, whereas in- have been shown to play an important role in
fection with variola minor resulted in less the resolution of orf virus infection.15 Using
than 1% fatality. Like ectromelia virus, vari- CD4+ and CD8+ T-lymphocyte depleted
ola virus has an extremely narrow host range, lambs infected with orf, CD4+ T-lympho-
infecting only humans. In addition, like ec- cyte depleted lambs were found to be unable
tromelia virus, variola virus causes a sys- to clear the virus, whereas CD8+ T-cell de-
temic infection. Infection occurs primarily pleted lambs were able to resolve the infec-
through the respiratory route (excretions of tion. Specific antibody titers have also been
the mouth and nose). The virus then repli- linked to the resolution of virus infection.
cates and spreads into the bloodstream, caus-
ing a primary viremia. After infecting and
replicating in many organs of the body, high G. Molluscum Contagiosum Virus
levels of variola virus are again shed into the Infection of Humans
blood, resulting in a secondary viremia. The
characteristic skin rash appears several days Molluscum contagiosum virus (MCV),
into the illness and progresses to a severe like variola virus, exclusively infects humans.
ulcerating rash that covers the body in about Unlike variola virus, MCV causes very small
10 days. Thanks to an aggressive vaccina- painless benign self-limiting lesions on
tion campaign organized by the World Health healthy individuals and only causes prob-
Organization (WHO), variola virus was eradi- lems in children and immunocompromised

155
adults (often disfiguring AIDS patients). In of wild voles and mice.22 Humans are ex-
addition to venereal transmission and direct posed to cowpox through contact with in-
contact with lesions, MCV is also believed fected udders, or more commonly through
to spread indirectly by tattooing, clothes, contact with infected domestic cats. Domes-
and bath towels. MCV is found worldwide, tic cats, exposed while hunting rodents, have
and studies done in Australia indicate that been shown to be highly susceptible to in-
23% of the overall population and 39% of fection. In fact, a study where young cats
adults over 50 years of age are seropositive, were infected with cowpox virus demon-
indicating that exposure is prevalent.16 Stud- strated an explosive disseminating disease
ies involving inoculation of volunteers dem- with 100% fatality.23,24 Therefore, it is felt
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

onstrate that MCV has an average incuba- that domestic cats act as intermediate hosts
tion period of 2 to 7 weeks. Although lesions in the circulation of CPV. The human dis-
usually regress within 3 months in healthy ease is generally mild zoonotic disease, al-
individuals, lesions have persisted for up to though severe, sometimes fatal (usually in
5 years in immunocompromised patients, immunocompromised patients) cases have
with reinfection occurring occasionally. The been reported.25 Lesions are often on the
treatment of MCV lesions is through a vari- face, hand, or neck, but occasionally gener-
ety of destructive techniques applied locally, alized possibly due to transfer from develop-
such as cryotherapy, none of which seem to ing lesion(s) on the hand. Shortly after infec-
work in patients with AIDS.17 MCV infection tion, the lesion(s) appears brownish and
is most noted for the lack of inflammation, becomes very painful after about a week.
despite high virus titers, in the lesion. Lesions Toward the end of the second week, the
For personal use only.

may persist for months without a sign of im- lesion ulcerates and crusts over. Although
mune cell infiltration. Studies have confirmed there is sometimes systemic involvement
the lack of T-lymphocytes and NK cells at the (usually in immunocompromised patients),
erupted lesion site.18,19 Research into MCV is the lesion usually heals within 3 to 8 weeks
difficult due to its inability to replicate in without scaring.26
tissue culture cells or animals. Recently, how-
ever, extended persistence of MCV DNA and
the expression of late proteins in an in vitro I. Lumpy Skin Disease Virus
cell culture has been demonstrated.20 Infection of Cattle

Lumpy skin disease virus (LSDV) ex-


H. Cowpox Virus Infection of Cattle, hibits a narrow host range, only infecting
Cats, and Humans cattle, most exclusively in Africa, with only
one outside case being reported in the Middle
Cowpox virus (CPV), despite its name, East.27 The disease causes hide damage, de-
very rarely infects cattle causing a mild pus- creased milk yield, infertility, poor growth,
tular disease of the teats and udders. Cow- as well as cattle death, resulting in major
pox has a very broad host range, in addition production loss and significant economic
to infecting cattle, cats, and humans, infec- problems. Lumpy skin disease is a subacute
tions in rats, zoo animals (lions, elephants, to acute disease characterized by either gen-
and anteaters), and gerbils have been re- eralized or localized nodular skin lesions
ported.21 The natural reservoir for the CPV and lymphadenopathy.28 The generalization
seems to be a small mammal; studies have of nodular skin lesions, due to systemic
demonstrated its presence in several species spread of LSDV, occurs in about 50% of

156
experimentally and naturally infected cattle losses to the poultry industry. In recent years
and correlates with high mortality. While vaccination by comb scarification, wing-web
dermal inoculation often results in local in- stick, or feather follicle immunization with
fection, intravenous inoculation of LSDV attenuated vaccine strains confers immunity
was shown to result in a much greater like- to FPV infection.36 In addition, better man-
lihood that a severe disseminating infection agement practices and improved hygienic
would take place.29 Vaccinations with at- conditions helped prevent the spread of the
tenuated strains, which cause only a mild disease. FPV spreads by two separate routes,
local infection, have been shown effective in each resulting in a different form of infec-
inducing strong immunity and controlling tion. Like SFV and LSDV, biting arthropod
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

the disease.30 Epidemiological, as well as vectors transmit FPV, resulting in the most
experimental, evidence has demonstrated that common form of the infection, the cutane-
LSDV is transmitted by arthropod vector, ous form. This form of the disease is often
either mosquitoes or sandflies.29,31 Therefore, characterized by mild focal skin lesions,
because of the increase in mosquito and bit- generally on unfeathered areas, sometimes
ing fly abundance during the wet season, leading to complete closure of the eyes, and
disease incidence also rises. may result in secondary bacterial infections.
The second disease form, transmitted through
the mucous membranes of the mouth, is much
J. Swinepox Virus Infection of Pigs more severe and disseminating. In this form
of the disease, the diphtheritic form, the in-
Swinepox virus (SPV) is found world- fection of the upper respiratory tract often
For personal use only.

wide, often infecting swine in both devel- results in suffocation due to closure of the
oped and underdeveloped countries. SPV has airway. Recently, recombinant FPV vaccines,
a narrow host range, infecting only porcine designed for the expression of foreign genes
species, and it causes a mild, benign, local in mammals (nonreplicating viral vector),
self-limiting infection. During infection, vi- have been studied extensively. Recombinant
rus can only be isolated from lesion site and FPV vaccines, encoding common HIV-1 an-
draining lymph nodes, confirming the lack tigens, have been studied for use as safe
of dissemination.32 When injected intrave- HIV-1 vaccine candidates.37
nously, a uniform distribution of lesions was
observed after an incubation period of be-
tween 10 and 14 days.33 The ultrastructure of III. HOST IMMUNE RESPONSE
SPV has been studied extensively in tissue
culture as well as the natural infection.34,35 The host immune response to poxvirus
infection is very powerful and multifaceted.
Additionally, the pathogenesis of localized
K. Fowlpox Virus Infection of and systemic infections varies dramatically,
Chickens depending on the host and poxvirus species.
EV infection of mice, for example, is often
Fowlpox virus (FPV) is found to infect severe and disseminated, but does not in-
primarily chickens and turkeys of all ages. duce an inflammatory response. MCV infec-
The disease stops egg production, stunts tion in humans also does not invoke an in-
growth of young birds, and causes blindness flammatory response, but remains localized.
and sometimes death, resulting in a major Other poxvirus infections result in inflam-
production loss and significant economic mation and can be either systemic or local,

157
for example, SFV is local, and FPV can be poxvirus species, and, most importantly, the
systemic. The common immune response host.
mounted against the poxvirus infection cov- To study the effect interferons have in
ers all arms of the immune system. Early in controlling poxvirus infection, knockout
the immune response, the innate immune mice, as well as anti-IFN antibody-treated
system is called into battle. Interferon, mice, have been infected with VV. Normal
complement, NK, and inflammatory cells mice have no problem clearing the infection;
rapidly try to keep the poxvirus under con- however, anti-IFN-treated mice were unable
trol until a more powerful acquired immune to clear the infection and died.38,39 Studies
response is mounted. Humoral and cell-me- using different IFN–/– and IFNR–/– knock-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

diated immunity is very important for virus out mice show similar results.40 However,
clearance and the prevention of reinfection. high levels of IFN in vivo do not always
Poxvirus-specific antibodies play many roles, correlate with the clearance of the infection.
including virus neutralization, opsonization, During EV infection of mice, high levels of
and ADCC. The cell-mediated immune re- IFN are present, and yet a lofty lethality
sponse, poxvirus-specific CTLs, is thought results.41 Interestingly, cell culture studies
to play the most important role in controlling show that the addition of exogenous IFN
poxvirus infection. results in abortive poxvirus infection. The
outcome seemed to be due in part to the
blocking of viral early mRNA translation.
A. Role of Interferon The effect of interferon block on poxvirus
infection in culture clearly varied depending
For personal use only.

Like other virus infections, interferons on the strain of virus used.


play a critical role in the immune response
against poxvirus infection. All three types of
interferon, alpha, produced by fibroblasts, B. Role of Complement
beta, produced by leukocytes, and gamma,
produced by lymphocytes and NK cells, ef- The role that complement plays in con-
fectively control poxvirus growth. Fortu- trolling poxvirus infection has been studied
nately, poxviruses are strong inducers of extensively. The complement system is com-
interferon production during animal infec- posed of over 30 serum proteins, some of
tion. Interferon then has two roles in control- which are regulatory in nature. It is a very
ling poxvirus infection. First, it warns neigh- important component of the innate immune
boring cells that a virus is present, resulting system and considered the first line of de-
in induction of an antiviral state, consisting fense against microbial infection. Although
of cell growth inhibition and the production three pathways of complement activation
of MHC antigens. Secondly, it causes acti- have been found, only two have been widely
vation of macrophages, NK cells, and cyto- studied for their role in blocking poxvirus
toxic T-lymphocytes. The cellular response infection: the classic pathway, initiated by
that ensues thereby helps clear virus-infected antibody fixation, and the alternative path-
cells. Although IFN is critical for the clear- way, initiated through the deposition of the
ance of poxvirus infections, interferon alone third component onto an activating surface.5
is not sufficient to protect the host from After the activation of either pathway,
severe disease. The role interferons play in proinflammatory chemotactic compounds are
poxvirus infection and pathogenesis signifi- released, resulting in leukocyte migration into
cantly depends on the route of inoculation, the area. The end result of complement acti-

158
vation is the formation of the membrane tional studies using C5+/C5– congenic BALB/c
attack complex (MAC), creating holes in the mice injected in the footpad with CPV
activating surface. As early as 1925, serum showed that C5 deficient mice exhibited a
neutralization of vaccinia virus was found to significantly greater swelling response that
be the result of heat-labile serum proteins, persisted longer and also showed signs of
later determined to be complement. How- hemorrhage, edema, and ulceration.48 Sev-
ever, no studies have been able to demon- eral studies have suggested that the role of
strate that complement alone can inactivate complement in controlling poxvirus infec-
purified poxvirus virions. tion varies, depending on the virus strain and
The classic pathway of complement ac- host being studied.
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

tivation is initiated through the binding of


antibodies and has been found to control
poxvirus infection by many mechanisms, C. Role of Inflammatory Cells
the first of which is virolysis. Once anti-
bodies are generated against the virus, se- The inflammatory response to poxvirus
rum-induced lysis of the virus occurs infection varies, depending on the species
through the formation of the MAC.42 Al- and host. Lesions induced by VV, SFV, and
ternatively, in the absence of lysis, poxvi- FPV infection, for example, are character-
rus neutralization has been found to take ized by leukocyte infiltration, swelling, and
place through the build up of complement redness. Amazingly, however, no inflamma-
components on the surface of the virus, tory response is seen in EV or MCV infec-
possibly blocking cell attachment. 43 tion. This is believed to be the result of viral
For personal use only.

Complement activation results in leuko- immune modulation. The role that inflam-
cyte migration and C3b fixation to the mation and inflammatory cells play in clear-
poxvirus surface, which could lead to an ing poxvirus infection has been studied for
increased phagocytosis of extracellular many years. It is clear that inflammatory
poxvirus through the binding of the C3b cells play an important role in controlling
receptor.44 Finally, poxvirus infection may poxvirus infection through a number of dif-
be controlled by complement through the ferent mechanisms. The first activity known
lysis of infected cells.45 to contribute to poxvirus depletion is phago-
The alternative pathway of complement cytosis. Inflammatory cells are able to ph-
becomes activated through the deposition of agocytize and destroy poxvirus virions that
C3 to an activating surface in the absence of are either antibody coated through binding
antibody. Again, this pathway results in the of the Fc receptor, or C3b coated, through
production of chemotactic compounds, as binding of the C3b receptor.49 Another way
well as the formation of the MAC. Cell cul- inflammatory cells are able to limit a poxvi-
ture studies of VV and FPV infection, dem- rus infection is through the release of oxy-
onstrated a time-dependent deposition of C3 gen-dependent and -independent antimicro-
onto the surface of infected cells.46 This C3 bial products.50 Finally, inflammatory cells
deposition correlated with a decrease in vi- are able to release interferon and other
rus yield, suggesting that the alternative path- cytokines, resulting in induction an antiviral
way of complement activation was control- state in local cells, as well as recruitment of
ling the infection. Additionally, the depletion new leukocytes into the area. In addition to
of C3 prior to infection of 3-day-old chick- the helpful effects contributed by leukocytes,
ens with FPV resulted in an increase in virus they have also been suspected of disseminat-
yield, leading to 100% mortality.47 Addi- ing virus in infections like that caused by

159
EV. This was evidenced by the recovery of ment-mediated lysis or opsonization, result-
EV from leukocytes present in the blood and ing in phagacytosis via the Fc receptor.
thoracic lymphatic duct. Antibodies can also bind the infected host
Experimental evidence shows that the cell, leading to antibody-dependent cell-
inflammatory response alone is not enough mediated cytotoxicity (ADCC). Despite the
to clear a poxvirus infection, without assis- numerous mechanisms created by specific
tance from an acquired immune response.51 antibodies to destroy poxviruses, many stud-
CPV lacking the gene encoding an inflam- ies have shown that antibody-mediated
mation modulating protein (serpin) showed mechanisms are unable to protect the host
a stronger inflammatory response in the CAM during a primary infection.
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

model and did not lead to a reduction of Studies of children who received a pas-
infection. This showed that the inflamma- sive immunization of gamma-globulin, fol-
tory response could not single-handedly halt lowing complications caused by vaccination
infection. However, studies have shown that against smallpox, confirmed that antibody
an inflammatory response is required to slow had no effect in reducing virus titers. In ad-
the progression of the poxvirus infection. dition, all patients with agammaglobuline-
One study using macrophage-depleted mice mia were found to develop protective immu-
(using carrageenan) demonstrated that after nity against smallpox following vaccination.
infection with a nonlethal dose of EV, an Even stronger evidence comes from the fact
increased level of virus was observed, re- that many patients who have succumbed to
sulting in death of the mice.52 Interestingly, complications from the smallpox vaccine
if macrophages were depleted 3 days after have had antivaccinia antibodies present in
For personal use only.

EV infection, virus titers were still high, their serum.53 This was confirmed with SFV
even though normal levels of CTLs were studies in rabbits; although neutralizing an-
observed; this demonstrated the importance tibody was detected in the serum of rabbits
of macrophages later in the infection pro- infected with SFV, tumor growth continued.
cess. It is clear that the inflammatory re- Additionally, passive immunization of in-
sponse, although essential for recovery, is fected immune compromised adult rabbits
not enough to control poxvirus infection with- with neutralizing antibodies to SFV also had
out the help of other host defense mecha- no effect on tumor growth. However, after
nisms. recovery from a primary poxvirus infection,
the host often develops antibodies respon-
sible for long-lasting immunity. The studies
D. Role of Antibody and ADCC looking at the ability of specific antibodies
generated during a primary infection to pre-
The effect that virus–specific antibodies vent a secondary infection have come with
have on poxvirus infections have been in- mixed results. Depending on the host, virus
vestigated quite extensively. Specific anti- species, infectious dose, and route of infec-
bodies have been found to be important in a tion, the virus-specific antibodies may or
number of mechanisms used to protect the may not be protective. Overall, it seems that
host. In addition to preventing virus poxvirus-specific antibodies are not as im-
uncoating, antibodies can directly bind to portant for recovery from a primary infec-
the poxvirus, causing aggregation and ob- tion as they are for the prevention of a sec-
structing viral adsorption and internalization ondary infection.
into the cell. Furthermore, antibodies can A subset of lymphocyte has been found
bind to the poxvirus, allowing for comple- to possess the ability to lyse antibody bound

160
virus-infected cells in a non-MHC-restricted EV and VV.57 Long before CTL activity was
manner.54 Because this cellular activity is detected (day 6), NK cell-mediated cytolysis
dependent on cell-bound antibody, it is very was observed. Additional studies have sug-
specific, but it is not present early in a pri- gested that over 50% of VV infected cell
mary infection. This process, known as lysis observed in hamsters was due to NK
ADCC, has been demonstrated in many stud- cells, not CTLs.58 Most surprisingly, how-
ies as being an important antibody and cell- ever, are the studies showing that mice strains
mediated mechanism used to combat sec- resistant to EV infection have higher levels
ondary virus infection. The addition of of NK cell-mediated lysis.59 In addition, they
anti-VV immune serum to VV-infected cells also showed that depletion of NK cells from
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

has been shown to enhance this subset of mice strains resistant to EV infection re-
lymphocyte’s ability to selectively destroy sulted in high virus titers and severe disease.
infected cells, without harming uninfected Studies later suggested that it was not the
cells.55 Like other immune mechanisms, the loss of NK cell-mediated cytolysis that gen-
effectiveness of ADDC in controlling pox- erated these results, but the loss of IFN pro-
virus infection depends on the poxvirus spe- duced by the NK cells, reducing the stimu-
cies and host.5 It is clear, however, that gen- lation of other antiviral mechanisms.60 Thus
erally ADCC is important for the repression far, the studies have suggested that NK cells
and elimination of poxvirus during reinfec- play an important role early (first 3 days) in
tion. the poxvirus infection, but cannot control
the spread and development of the infection
on their own.
For personal use only.

E. Role of NK Cells

The non-MHC restricted, nonspecific F. Role of CTLs


cytolytic activity of NK cells can be seen long
before the development of virus-specific While many components of the host
CTLs. NK cells, which appear as large granu- immune response are required for control-
lar lymphocytes, are found at the area of virus ling poxvirus infections, it is generally felt
infection as part of the inflammatory response, that the most important role is played by the
and part of the innate immune response. After virus-specific CTL. The CTL (CD8+) is an
the release of interferon by infiltrating leuko- important part of the acquired cell-mediated
cytes, NK cells become activated and begin immune response. It is MHC class I restricted,
lysing virus-infected target cells by the re- specifically recognizing virus-infected cells,
lease of perforin and granzymes.56 NK cells destroying them before virus maturation, and
are able to nonspecifically discriminate be- thereby reduces the spread of infection. As
tween normal cells and virus-infected cells, CTLs are part of the acquired immune re-
which do not express adequate amounts of sponse, they are not present early in the pri-
major histocompatibility complex (MHC) mary infection. They are important for clear-
class I molecules on their surface. Studies ance later in the infection, as well as the
have consistently demonstrated the impor- rapid response and virus elimination after
tance of NK cells early in the immune re- reinfection.
sponse to poxvirus infections. Mouse studies of poxvirus-specific
The early activity of NK cells during CTLs, generated during primary infection,
poxvirus infection was shown in mice in- vary slightly in their results, depending on
fected intracranially with sublethal doses of the strain used, route of inoculation, and

161
virus dose.61 Primary CTL responses were used to describe virally encoded proteins
greatest with high doses of virus and intrave- that are secreted from the infected host cell.
nous inoculation.62 Studies that measured the Most virokines are powerful viral immune
CTL response in mice infected with EV modulating proteins created by capturing and
showed that virus-specific killing of target modifying genes responsible for regulating
cells peak around the sixth day of infec- the host immune response. Therefore, these
tion.63 After reinfection, the CTL response viral proteins share many structural and func-
was shown to be eight times greater and was tional features of that of the host proteins.
detected earlier, around the fourth day of However, because there is limited room
infection.64 These studies illustrate the clas- within the viral genome, only those genes
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

sic pattern for a learned CTL immune re- very useful to the virus are maintained and
sponse to primary virus infections, and the constantly adapted to make them smaller
faster, more powerful memory response to and more potent. There have been virokines
reinfection. The importance of CTLs in vi- discovered and shown to modulate many
rus clearance and recovery was demonstrated important aspects of the immune response.
many years ago in EV-infected mice. Due to Many cytokine (IL-6, IL-10) and cytokine
the clear correlation between the increase in receptor (TNFR, INFγR, IL-1βR) homologs
CTL activity and virus clearance and host (also referred to as viroceptors or virokines
recovery, studies concluded that CTLs were that mimic receptors) have been shown to
responsible for protection.61 significantly disrupt cytokine-signaling path-
ways. This often results in the masking of
the host immune system from the viral infec-
For personal use only.

IV. VIRAL COUNTER RESPONSE tion, allowing the virus to grow to high ti-
ters. Chemokine homologs and chemokine
In order to defend themselves from the inhibitors (some of which are chemokine re-
many arms of the immune system, poxvi- ceptor homologs) were found to have a dra-
ruses must constantly evolve new, more matic effect on leukocyte chemotaxis. These
powerful methods of modulating the host virokines have been shown to either help the
immune response. Being large DNA viruses, virus evade immune detection, in some cases
poxviruses have enough room to encode by blocking the influx of leukocytes into the
proteins not directly involved in replication. area of infection, or in other cases, actually
Therefore, they often encode a repertoire of cause the recruitment of leukocytes to in-
proteins involved in habitat formation and crease the pool of new host cells to allow for
immune evasion (see Figure 2). These pro- better dissemination. Chemokine inhibitors
teins cover a wide range of activities, re- have been identified and shown to block cel-
flecting the scope and complexity of the lular influx into the areas of infection. There
immune response that they counter in their are also virokines that block the most impor-
host. The survival of the virus depends on its tant aspect of the innate immune system, the
ability to create and perfect strategies to complement system. These proteins are very
conceal itself from this much larger and more powerful; they not only prevent the formation
sophisticated opponent. The creativity and of the membrane attack complex (MAC), but
potency of the resulting proteins are fasci- also block the complement cascade at mul-
nating and cover both intracellular and ex- tiple sites upstream to block the formation of
tracellular processes. Thus far, most of the proinflammatory factors. Therefore, they are
proteins that have been discovered are extra- also able to greatly reduce the migration of
cellular. The term virokine was coined and leukocytes into the area of viral infection.

162
In addition to those proteins encoded by (MCP), decay-accelerating factor (DAF),
poxviruses that are secreted from the host complement receptor one (CR1), and is struc-
cell, there are also proteins that act intracel- turally most similar to human complement 4b
lularly. The most studied group of intracel- binding protein (C4b-BP). Purified bioactive
lular poxvirus immune modulating proteins VCP has been shown to inhibit both the clas-
are the serine proteinase inhibitors (serpins).65 sic and alternative pathways of complement
Many poxviruses have been found to encode activation through its ability to bind C3 and
serpins that modulate programmed cell death, C4 and act as a cofactor for factor I cleavage
apoptosis. By blocking the cysteine protein- of C3b and C4b.69 VCP, secreted from virally
ase interleukin-1β-converting enzyme (ICE), infected cells, is believed to protect the cell
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

and the serine proteinase granzyme B, the and the released viral progeny from host
cowpox cytokine response modifier A complement attack. In addition, by blocking
(CrmA), vaccinia virus SPI-2, and myxoma complement activation at multiple sites, there
virus Serp-2 all have been shown to inhibit is a large reduction of C3a, C4a, and C5a
apoptosis.66 By blocking these intracellular proinflammatory chemotactic factors, result-
enzymes, these poxvirus serine protease in- ing in reduced cellular influx and inflamma-
hibitor (serpins) proteins are able to prevent tion.
TNF- and IFN-induced, as well as T-lym- VCP has also been shown to possess the
phocyte-mediated Fas-induced apoptosis.67 ability to bind heparin, as well as heparan
By protecting the infected cell from pro- sulfate proteoglycans (heparin-like mol-
grammed cell death, the virus is able to rep- ecules) on the surface of human endothelial
licate to higher titers and continue host eva- cells. Because of this activity, VCP has been
For personal use only.

sion. shown to exhibit many properties that all


other complement control proteins are lack-
ing. In vivo studies have indicated that FITC
A. Complement Regulatory Proteins labeled VCP, when injected intravenously,
becomes localized in the endothelium. This
Many poxviruses encode proteins that very interesting property of VCP has been
mimic components of the host complement shown in vitro to reduce chemotactic migra-
system in order to block the activation of tion of leukocytes in the presence and ab-
complement and thereby modulate the inflam- sence of the chemokine MIP-1α.70 In addi-
matory response mounted by the host.68 The tion, VCP was able to inhibit antibody
vaccinia virus complement control protein binding to MHC class I molecules on human
(VCP) was the first soluble microbial protein endothelial cells.71 This suggested that VCP
shown to have complement binding activity could interfere with molecular interactions
and postulated to have a role in evasion of with infected cells and possibly prevent an-
host complement. In the years since its dis- tibody-dependent cell-mediated cytotoxicity
covery, VCP has become one of the most (ADCC) as well as other cytotoxic cell inter-
well-studied and best-characterized virokine. actions with target cells. Amazingly, VCP
Structurally, it is composed of four comple- was shown to block interactions between pig
ment control protein (CCP) modules (also aortic endothelial cells (PAECs) and naïve
called short consensus repeats), making it and neutrophils and natural killer cells, prevent-
other homologous proteins members of the ing cell killing.72 In addition, cell culture
regulators of complement activation (RCA) studies demonstrate that VCP blocks comple-
superfamily. VCP exhibits functional simi- ment-mediated killing of PAECs by human
larity to factor H, membrane cofactor protein serum in a dose-dependent manner.

163
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15
For personal use only.

164
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15
For personal use only.

FIGURE 2. Poxvirus immune modulation. (1) Poxvirus binds the host cell, allowing for core entry and early gene transcription to initiate. At this time, many viral
proteins are produced to interfere with the host’s immune response in order to establish a suitable habitat for optimal growth. (2) Vaccinia virus complement control
protein (VCP) is produced and secreted from infected cells. VCP can block the activation of complement through its ability to bind C3b and C4b complement
components. VCP can also bind heparan sulfate proteoglycans on the cell surface, blocking many molecular and cellular interactions with the cell, like chemokine
and antibody binding. (3) Poxviruses also produce a soluble interleukin one receptor homolog (vIL-1βR) that is secreted from the cell, binding up soluble IL-1β
before it has a chance to reach the cell’s receptor. (4) Chemokine binding proteins, such as T1 and T7, inhibit chemokine binding to heparan sulfate proteoglycans
on the cell surface. (5) Soluble viral interleukin ten (vIL-10) is secreted from the cell, binds to IL-10 receptors on inflammatory cells, blocking cell activation. (6)
Soluble poxvirus interferon gamma receptor homolog (vIFN-γR) is also secreted from infected cells, binding and preventing host IFN-γ from binding the cellular
receptor. (7) Another poxvirus early protein that is responsible for immune modulation, soluble tumor necrosis factor receptor (vTNFR) homolog, binds host TNF
and prevents its interaction with the cellular receptor. (8) Poxvirus interleukin eighteen binding protein (vIL-18BP) is secreted form the infected host cell and binds
soluble IL-18, inhibiting its proinflammatory functions. (9) Orf virus encoded granulocyte-macrophage colony stimulating factor-inhibitory factor (GIF) is secreted
and binds to granulocyte-macrophage colony-stimulating factor (GM-CSF), blocking its ability to stimulate the production and secretion of IFNα. (10) Myxoma
virus SERP-1, which is the only secreted poxvirus serpin, blocks several serine proteases, leading to reduction of inflammation. (11) The intracellular poxvirus
serine protease inhibitor proteins, such as SPI-2, serp-2, and crmA, block the conversion of pre-caspase to active caspase one, thereby blocking its ability to
convert pro-IL-1β to IL-1β and the cell’s ability to undergo programmed cell death.

165
B. Cytokine Homologs C. Cytokine Receptor Homologs
(Viroceptors)
The parapoxvirus Orf, which often infects
sheep and goats (sometimes humans), very Many members of the poxvirus family,
recently has been shown to encode a viral including myxoma, vaccinia, and cowpox,
homolog of IL-10 (vIL-10).73 This protein encode homologs of the tumor necrosis fac-
shows the greatest degree of homology, 80%, tor receptor (TNFR). This viral tumor necro-
to sheep IL-10. Endogenous IL-10 plays many sis factor receptor (vTNFR) was originally
important roles in down-regulating the immune described in myxoma, where it is known as
system. One very important function is to sup- T2, and Shope fibroma viruses (SFV), and
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

press the activation of macrophages, thereby the term viroceptor was proposed to describe
blocking their ability to act as accessory cells them.76 TNF is a very important cytokine for
to activate T-cells or natural killer (NK) cells the early response to viral infection. It is a
and secrete cytokines. Thus, IL-10 acts as a very powerful proinflammatory agent, trig-
potent antiinflammatory agent and a powerful gering numerous immunological events, in-
immunosuppressor. Because of this activity, it cluding apoptosis of virally infected cells. In
is believed that the Orf virus produces vIL-10 order to block the effects of this powerful
in order to inhibit the expression of various cytokine, these poxviruses have captured and
cytokines that regulate the immune response modified the host gene for TNFR so that it
against viral infection, suppressing the activa- may be secreted in large amounts from the
tion of macrophages. The benefits of vIL-10 infected cell. The vTNFR can bind soluble
production for the virus are thereby to mask TNF present in the nearby area, preventing
For personal use only.

viral infection from the immune system long its binding to the cellular TNFR, thereby
enough to grow to high titers. blocking the action of this cytokine. Many
Very recently, a new virokine, having poxviruses encode several copies of this gene;
homology to human interleukin 18 binding cowpox, for example, contains three sepa-
protein (hIL-18BP), was uncovered in mol- rate copies, showing its importance to viral
luscum contagiosum virus (MCV).74 Since replication.77 In addition, when vTNFR (T2)
this time, it has also been found in other was deleted from myxoma virus the viru-
poxviruses, including ectromelia, vaccinia, lence was reduced significantly. Interestingly,
and cowpox.75 IL-18 is a proinflammatory vTNFRs show a great deal of host specific-
cytokine important in activating natural killer ity. Myxoma virus vTNFR, T2, will only
(NK) cells, inducing a T-helper 1 response, bind to rabbit TNF (not at all to human),
and inducing the production of interferon whereas the vTNFR encoded by vaccinia
gamma (IFNγ). The MCV vIL-18BP has been and cowpox virus will bind human, mouse,
shown to bind to both human and murine IL-18 and rat TNF effectively.
and inhibit the production of IFNγ in a dose- Many poxviruses, including variola, ec-
dependent manner.74 The deletion of the gene tromelia, vaccinia, cowpox, and camelpox,
encoding vIL-18BP in vaccinia was shown also encode an additional interesting cytokine
to dramatically attenuate the virus.75 In addi- receptor homolog, viral interferon gamma
tion, the ectromelia virus vIL-18BP was receptor (vINFγR). These proteins greatly
shown to block IL-18-induced NF-κB acti- resemble the N-terminal, ligand binding (ex-
vation and induction of IFNγ. Therefore, this tracellular domain) region of their mamma-
strategy of viral evasion also acts to indi- lian counterparts. As with vTNFR, these
rectly block the strong antiviral effects of cytokine receptor homologs are also very
IFNγ. species specific. The first and most exten-

166
sively studied of these proteins comes from trum of inflammatory responses. NK cells
the myxoma virus and is known as T7.78 T7 and B-cells proliferate in response to IL-1,
is a 37-kDa N-linked glycoprotein that is the neutrophils degranulate, and T-cells produce
most abundantly secreted protein of myxoma IL-2 and proliferate.
virus. Exhibiting great species specificity, Another very recently identified virokine
T7 was shown to bind and block the effects was found encoded by the parapoxvirus, Orf.82
of rabbit INFγ, but had no effect in blocking The Orf virus produces a secreted protein,
human or murine INFγ. In contrast, vaccinia termed granulocyte-macrophage colony stimu-
virus, which has a broad host range, can bind lating factor-inhibitory factor (GIF), which was
and block a number of mammalian INFγs. shown to bind to and block the activity of GM-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

The importance of this encoded protein to CSF, as well as IL-2. Both of these cytokines
myxoma virus virulence is evident, not only are very important in host antiviral immunity;
because it is the most abundantly secreted IL-2 is essential for its role in the activation of
protein, but also because deletion of the gene T-cells, and GM-CSF for its ability to stimu-
eliminates the 99% mortality rate exhibited late the production and secretion of IFNα. This
by the virus, and all infected rabbits recover. remarkable virokine plays a very novel role in
Therefore, the T7 protein represents the major modulation of host defense as a means of pre-
virulence factor of myxoma virus. The im- serving viral habitat.
portance of this protein to viral virulence is
due to INFγ’s powerful antiviral effects. INFγ
serves numerous functions, including switch- D. Chemokine Homologs
ing on many intracellular antiviral activities,
For personal use only.

and giving the signal to nearby cells that a Many poxviruses encode proteins that
viral infection is occurring. Another cytokine inhibit chemokines; the CC (β) chemokine
receptor homolog, produced most notably homolog encoded by the molluscipoxvirus
by the orthopox members vaccinia and cow- molluscum contagiosum (MCV), designated
pox, is viral interleukin 1 receptor (vIL-1R). as molluscum contagiosum chemokine ho-
This 33-kDa protein was first identified in molog (MCCH) or MC148, was discovered
vaccinia virus and found to have the ability only recently.83 This human disease causing
to bind murine IL-1β in 1992.79 A similar virus presumably utilizes this virokine to
open reading frame was later identified in block leukocyte trafficking to the site of in-
cowpox virus and the protein product also fection. MC148 is highly homologous to
found to bind murine IL-1β. Interestingly, members of the CC or β chemokine family;
supernatants taken from vaccinia virus in- however, it is truncated in the N-terminus of
fected cells were found to inhibit IL-1-in- the protein. The N-terminal portion of the
duced murine lymphocyte proliferation. In chemokine is required for proper binding
addition, recombinant vaccinia, unable to and activation of the cell.83 Therefore, the
express vIL-1R, showed remarkable attenu- MC148 chemokine homolog of MCV acts
ation compared with the wild type in vivo.80 by binding to the chemokine receptor, iden-
This demonstrates how the protein plays a tified as being specifically CCR8, thereby
critical role in the pathology of vaccinia vi- blocking endogenous chemokine binding.
rus infection. The importance of IL-1 in The overall result of this protein is reduced
combating viral infection is large and was cellular influx and therefore reduced inflam-
demonstrated many years ago.81 The expres- mation. This protein seems to be geared to-
sion of IL-1 is induced early on in many ward specifically blocking monocyte infil-
viral infections and results in a broad spec- tration and dendritic cell function.

167
E. Chemokine Inhibitors pox cytokine response modifier A (CrmA),
vaccinia virus SPI-2, and myxoma virus Serp-2
Many poxviruses encode a protein that have all been shown to inhibit apoptosis
blocks the effects of chemokines. Collectively, through the ability to block both the cysteine
these virokines have been termed the T1/35kDa proteinase interleukin-1β-converting enzyme
family, all of which possess eight conserved (ICE) and the serine proteinase granzyme
cysteine residues and are shown to interact B.66,86,87 By blocking these intracellular en-
with members of the CC and/or CXC classes zymes, these poxvirus serine protease in-
of chemokines.84 The myxoma virus T1 and hibitor (serpins) proteins are able to prevent
T7 proteins are the most widely studied mem- TNF and IFN-induced, as well as, T-lym-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

bers of this group. T1 shows no homology to phocyte-mediated (Ca+2-independent) Fas-


any host chemokine or chemokine receptor induced apoptosis.66,88 Therefore, these pro-
and binds strongly (specifically) to members teins prevent destruction of infected cells,
of the CC class of chemokines, but only weakly even if they are recognized as being infected,
(nonspecifically) to CXC chemokines.65 Be- thus maintaining a habitat conducive to vi-
cause of this ability, T1 has been shown to rus replication.
block human monocyte migration in the pres- Many members of the poxvirus family
ence of MIP-1α in vitro. The deletion of the encode proteins that inhibit serine proteases
coding sequence for T1 resulted in a pronounced intracellularly, but only myxoma virus en-
increase in infiltrating monocytes and mac- codes one that acts as a serine protease inhibi-
rophages into the site of infection in vivo.84 tor (serpin) and secreted extracellularly. First
However, the loss of T1 had no significant identified in 1990, this secreted glycoprotein,
For personal use only.

effect on mortality, indicating that this gene Serp-1, has structural features that place it in
does not play a critical role in the pathogenesis the serpin family of serine protease inhibi-
of the infection. T7, on the other hand, has tors.89 Before its discovery, intracellular
been shown to inhibit the activity of members serpins were found in vaccinia virus.90 Serpins
of C, CC, and CXC classes of chemokines in have many functions in maintaining homeo-
a nonspecies-specific manner. This protein, stasis of eukaryotic systems, from comple-
which is most noted for its anti-IFNγ activity, ment activation and blood clotting to the in-
has been shown to nonspecifically bind to the flammatory response. In addition, mutations
heparan sulfate proteoglycan binding domain in serpins have been linked to over 90 human
of the chemokine, preventing gradient forma- diseases. Given the importance of these pro-
tion along the vascular wall, and inhibiting teins, many pathogens have evolved to carry
chemotaxis.85 The addition of excess soluble their own serpins to disrupt host processes.
heparin completely prevents this activity of the Myxoma virus Serp-1, for example, has been
T7 protein, supporting this hypothesis. Because shown in vivo to weaken the inflammatory
the T7 protein also exhibits no homology to response to viral infection. When this gene is
any known chemokine, this strategy of deleted from the virus, a much greater inflam-
chemokine inhibition is quit novel. matory process is observed and the infection
is resolved much more rapidly. Purified re-
combinant Serp-1 has been shown to bind a
F. Serine Protease Inhibitors variety of proteases, including plasmin, throm-
bin, tissue-type plasminogen activator (tPA),
Several poxviruses have been shown to and urokinase-type plasminogen activator
encode proteins that are able to prevent (uPA), although the exact target has not been
apoptosis, programmed cell death. The cow- found.91 This protein acts as a pseudosubstrate

168
for these serine proteases, forming a stable the virus strains (see Figure 3). The degree of
complex, preventing the protease from bind- inflammation and cellular infiltration can be
ing its true substrate. Due to the role of Serp-1 both quantitatively and qualitatively analyzed.
in modulating inflammation, recently it was This allows for better characterization of mu-
tested in a restenosis model system to evalu- tant strains and results in a better understand-
ate its therapeutic value. ing of genes associated with poxvirus viru-
lence (see Table 2). Understanding poxvirus
virulence aids in the production of better, more
V. ANIMAL MODELS FOR THE attenuated, vaccine strains, in addition to pox-
STUDY OF POXVIRUS INFECTIONS virus vectors for use in new vaccines, in hopes
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

of reducing complications associated with tra-


Several animal models have been devel- ditional vaccines. Several important models
oped and used to study various poxvirus dis- that have been used extensively in poxvirus
eases and poxvirus mutants to better our un- research are discussed in greater detail.
derstanding of the infection processes and
virulence genes. Animal infection models are
important for exploring new treatment strate- A. Chicken Embryo Chorioallantoic
gies and are used to test the efficacies of anti- Membrane (CAM) Model
viral drugs. Models have been developed to
study the inflammatory response generated by The Chicken embryo chorioallantoic mem-
poxvirus infection, as well as the virulence of brane (CAM) model has been used extensively
For personal use only.

FIGURE 3. Poxvirus infection models. (A) Footpad of normal mouse. (B) Swollen footpad of a mouse several
days after injection of CPV. The degree of inflammation can be measured. (C) Red pocks formed on CAM of
12-day-old chicken embryo 3 to 4 days after infection with CPV, showing no inflammatory response. (D) White
pocks formed on CAM of 12-day-old chicken embryo 3 to 4 days after infection with a mutant strain of CPV,
showing a massive inflammatory response.96 (E) Mouse intranasally infected with mutant VV, showing little
signs of infection. (F) Mouse intranasally infected with wild-type VV, showing significant signs (ruffled fur) of
pathology.

169
TABLE 2
Poxvirus Animal Models for the Study of Poxvirus Infections. Many of the Experiments
Utilizing Various Animal Models to Study Poxvirus Infection Are Listed. Also Listed Are
the Poxvirus, or Mutant Poxvirus Tested and a Short Description of the Experimental
Model
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15
For personal use only.

170
to study pathogenic poxvirus genes and for the induced by poxvirus infection. Poxvirus is
diagnosis of poxvirus infection. The CAM of an injected into one of the hind footpads of an
11- to 12-day-old embryonated chicken egg is adult mouse or rat. The primary inflamma-
inoculated with virus and incubated for 3 days. tory response is then measured over several
The CAM is then removed and the color and days. In this model, unlike the CAM model,
morphology of the pock lesions is examined. a fully functional immune system is present,
This system is often used to study poxvirus and the inflammatory response can be calcu-
virulence genes and the presence or absence of lated through direct measurement of the
an inflammatory response. The CAM model is swollen footpad. Like the CAM model, how-
limited, however, because chicken embryos lack ever, it is very quick and easy to perform.
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

a fully developed immune system. Therefore, Cellular infiltrate is often examined micro-
this model does not truly represent the infection scopically from the footpad to better charac-
process of a poxvirus in vivo. terize the specific cell types. This model is
Experimentally, the CAM model has been used primarily to determine the role various
used to study poxvirus-encoded genes involved poxvirus genes have in regulating the in-
in inflammation modulation, characterize new flammatory response.
poxvirus strains and mutants, and analyze ge- Early experiments used the footpad
netic traits of recombinant poxviruses. Recom- model to assess the cell-mediated immune
binants between EV and VV were found to response to VV infection in rats.100 The ki-
possess pathogenicity markers from both parent netics of the cellular immune response was
poxviruses using the CAM model.92 Recombi- found to be in accordance with the regular
nant virus produced pocks that were large, hem- time course. Footpad swelling started 3 days
For personal use only.

orrhagic, and ulcerating, far more severe than after infection, peaked around day 6, and
that formed by the parents, indicating that patho- disappeared after 2 weeks. The repose was
genicity was enhanced in the recombinants. A comparable to that reported previously in
study of the cell proliferative response to VV mice. More recently, the footpad model was
growth factor (VGF) was also examined in the employed to determine the role that a poxvi-
CAM model.93 VGF was found to produce le- rus encoded complement regulating protein
sions that contained more proliferating cells and might have on inflammation.48 To better
more infectious virus progeny. Similar studies simulate an actual infection process, cow-
use the CAM to analyze the effect gene dele- pox virus (CPV) was injected into the foot-
tions have on the pock morphology and cellular pads of mice, the natural host. Wild-type and
infiltration.94–97 The deletion of genes encoding recombinant CPV without the gene encod-
inflammatory modulating proteins results in a ing the complement regulatory protein,
change of pock lesions from red to white, an termed the inflammation modulatory protein
indication a massive cellular inflammatory re- (IMP), was injected into the footpads of
sponse. Finally, the CAM is used to characterize mice.48,101 Additionally, C3 and MIP-1α
new poxviruses and poxvirus strains, using varia- knockout mice were injected with CPV and
tions in pock size, color, and morphology.98,99 CPV lacking IMP, and the footpad swelling
measured. Significantly greater footpad
swelling was observed in the mice injected
B. Footpad Model for Quantitative with CPV lacking IMP. The results con-
Analysis of Inflammation firmed the importance that IMP has in re-
ducing cellular infiltration by inhibiting
The footpad injection model was devel- complement activation, thereby reducing the
oped to assess the inflammatory response production of chemotactic factors.

171
C. Air Pouch Model for Qualitative define poxvirus virulence genes, most ex-
Assessment of Inflammation clusively in VV. The spontaneous deletion
mutant 6/2 of VV, which lacks 17 open read-
The subcutaneous air pouch injection ing frames (ORF), was injected intracrani-
model was developed to assess the precise ally to demonstrate a loss in virulence.104 An
cellular changes at the site of poxvirus infec- enormous loss in virulence was observed
tion. It has long been used to assess the compared to the wild-type VV strain. An
inflammatory response elicited by many additional study using the intracranial injec-
natural and synthetic agents, as well as the tion model showed a substantial reduction of
ability of various drugs to block this re- VV virulence when a single 13.8-kDa ORF
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

sponse. Recently, it has been proven to be a was deleted.105 The single protein encoded
valuable tool for examining the cellular in- by this ORF contributed much of the viru-
filtrate at the site of poxvirus infection. The lence demonstrated by wild-type strain of
model is simple; mice or rats are injected in VV. The overall goal of these studies are to
the back subcutaneously with 1 cc of air. identify mutant strains of VV that may be
Virus or some other material of interest is used for vaccination and gene therapy, with-
then injected into the air pouch. The air pouch out the complication risks traditionally asso-
is then harvested at different time points; the ciated with VV vaccine strains, such as
thin layer of connective tissue is spread across postvaccinial encephalitis.
a slide, stained (with May-Grunwald and
Giemsa), and the cellular infiltrate examined
microscopically. This model was used suc- E. Nude Mouse Model for
For personal use only.

cessfully to evaluate the importance IMP of Evaluation of Virulence


CPV has in limiting inflammatory cell
chemotaxis to the site of infection.101 Poxvirus virulence is also evaluated in
the nude (athymic) mouse model. The model
involves intraperitoneal injection of VV into
D. Intracranial Injection Model athymic mice, between 4 and 16 weeks of
age, and mortality is measured out to 60
The virulence of poxvirus has often been days. A dose-dependent mortality is observed
tested using the intracranial (LD50) injection with virulent strains of VV. This model has
model. A direct correlation between been used quite extensively to investigate
neurovirulence and virus propagation has the biological roles of various cytokines and
been shown previously with VV inoculated chemokines expressed by recombinant VV.
intracranially, higher lethality resulted form Nude mice are also used to study the viru-
greater virus yield.102,103 Weanling mice lence of mutant vaccinia virus for possible
(3-to 4-week-old male BALB/c) are gener- use as vaccine strains. Several deletion mu-
ally used for this model due to the greater tants of VV have been shown to have re-
ease in which the softer skull can be pen- duced virulence, when compared with the
etrated. Using a small-gauge needle, virus is wild-type strain, when injected intraperito-
injected intracranially into one of the cere- neally into nude mice.105 Other studies show
bral hemispheres of the mice. Long-term that expression of various cytokines, within
mortality can be observed or the mice can be recombinant VV, reduces or prevents infec-
sacrificed at various time points to deter- tion in nude mice. VV expressing human or
mine the virus titer in the brain tissue. Ex- mouse IL-2 was rapidly eliminated, result-
perimentally, this model has been used to ing in little or no mortality.106 A similar ef-

172
fect, although not to the same degree, was VI. ANIMAL MODELS FOR THE
seen when VV expressing IL-15 was in- STUDY OF POXVIRUS IMMUNE
jected into nude mice.107 The effect of IL-2 MODULATION
was not systemic, coinfecting with IL-2 ex-
pressing and virulent wild-type strains, re- As discussed earlier, poxviruses are well
sulted in significant lethality.108 In addition, noted for their ability to produce a repertoire
the effects seen by IL-2 were abolished when of immune modulating proteins. Several of
the mice were injected with monoclonal these proteins have been investigated further
antibody against IFNγ.109 This model has for their potential use as therapeutic agents
also been used to evaluate the effects that (see Table 3). These immune blocking pro-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

VV expressed cytokines and chemokine have teins have very powerful antiinflammatory
on the host cellular response to infection. properties that may be employed for treat-
The enhancement of NK cell activity was ment of many injuries or diseases that in-
attributed to many of the effects observed in volve excessive inflammation. Among other
these studies.107,110 things, poxvirus immunomodulatory proteins
have been explored for potential application
in transplantation, vascular injury, CNS in-
F. Intranasal Inoculation Model jury, sepsis, arthritis, and restenosis. Two
poxvirus-encoded proteins have been stud-
Intranasal inoculation of mice with ied quite extensively, VCP and Serp-1; the
CPV has been used as a model to study various animal models employed to evaluate
orthopoxvirus respiratory disease. Mice, 4 their therapeutic effectiveness is discussed
For personal use only.

to 6 weeks of age, are inoculated intrana- in greater detail.


sally with aerosols containing CPV.111 Re-
cently, this model has been used to study the
efficiencies of antipoxvirus drugs in hopes A. Xenotransplantation Model
of developing better treatment strategies for
the highly fatal respiratory disease often as- In order to test the ability of the vaccinia
sociated with human monkeypox virus in- virus complement control protein (VCP) to
fection.112 Severe disease, resulting in death, block the rejection of grafted tissue trans-
starts to take effect by day 7 in untreated planted between species, a suitable animal
mice. Mice develop pulmonary lesions, even- model was developed. Guinea pig-to-rat het-
tually involving most of the visceral pleura, erotopic heart transplantation was performed
and often show ulcerating lesions on the tail, using young guinea pigs and large adult rats.
indicating dissemination. Cidofovir, an ap- Guinea pig hearts were removed, prepared in
proved antiviral drug, has been shown to cold cardioplegic solution, and microsurgically
significantly reduce mortality using the in- transplanted into the neck of the rat. With all
tranasal inoculation model.113,114 Addition- other vessels of the donor heart ligated, the
ally, the intranasal inoculation model is used left jugular vein of the rat is sutured to the left
to characterize mutant strains of VV. Intra- pulmonary artery of the heart and the right
nasal inoculation of mice with wild-type VV carotid artery of the rat is sutured to the in-
resulted in a highly lethal disseminated in- nominate artery of the aorta. The clamps are
fection.115 However, mice intranasally in- released, blood enters and warms the donor
fected with VV lacking the thymidine kinase heart, and the heart spontaneously begins to
gene showed no lethality and a lack of dis- beat. Steady perfusion of the donor heart tis-
semination.116 sue with the rat’s blood, powered by the rat’s

173
TABLE 3
Poxvirus Animal Models for the Study of Therapeutic Poxvirus Immune Modulation:
Many of the Experiments Utilizing Various Animal Models to Study Poxvirus Immune
Modulation Are Listed. Also Listed Are the Poxvirus Proteins Tested and a Short
Description of the Experimental Model
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

heart, exposes the donor heart to the immune plaque development, several animal models
system of the rat. Because this model repre- were used.118 An aortic allograft model was
sents a xenotransplant, the donor heart is employed to investigate artheroscletotic plaque
For personal use only.

quickly rejected, between 15 and 20 min after formation following graft transplantation in
blood flow is established. This rejection oc- rats. A 1-cm section of lower abdominal aorta
curs very rapidly due to the presence of anti- was removed for the donor and sutured end to
bodies in the blood of the rat, which cross- end into the abdominal aorta of the recipient
react to sugar moieties on the endothelium of rat. After blood flow was established, a single
the guinea pig heart. This will then lead to the dose of Serp-1 or saline was injected intrave-
activation of complement and thereby the de- nously. The tissue was then harvested for
struction of the donor heart. Due to the comple- histological examination to determine the
ment inhibiting ability of VCP, as well as the short- and long-term effects. The single dose
other properties contributed by its ability to of Serp-1 was found to significantly reduce
bind heparin, its capacity to block or delay inflammatory cell infiltration and later plaque
this rejection process was examined. Intrave- formation and aortic occlusion.119 This ability
nous administration of VCP was found to to block artheroscletotic plaque formation was
delay xenograft rejection, by not only block- also tested in a rabbit model for balloon
ing complement activation, but also prevent- angioplasty. Cholesterol-fed rabbits were in-
ing antibody fixation to the donor endothe- fused with Serp-1 or saline shortly after being
lium.117 subjected to balloon angioplasty. The injury
site tissue was removed at different time points
for histological examination. Again, Serp-1
B. Allograft and Restenosis Models: was found to dramatically reduce cellular in-
vasion and plaque formation at the site of
To test the therapeutic ability of Serp-1, injury.120
the MV secreted antiinflammatory serine pro- The M-T7 protein of MV, which is able
teinase inhibitor, to suppress artherosclerotic to inhibit IFN-γ and block chemokines, was

174
also tested for its ability to suppress injury, resulted in a significant reduction of
artherosclerotic plaque development follow- inflammatory cell infiltration and comple-
ing balloon angioplasty.121 Using both rat ment activation.123 However, these results,
and rabbit animal models, purified M-T7 or which demonstrate a general reduction in
saline was infused into the area after the inflammation, do not necessarily imply that
injury. In both models, histological analysis neuronal sparing and/or cognitive recovery
after 1 month revealed a significant reduc- will occur. Therefore, to assess whether ad-
tion in arterial hyperplasia. In addition, an ministration of VCP at the site of brain in-
early inhibition of inflammatory cell inva- jury can lead to a functional recovery, the
sion into the area of the injury was observed. special memory of brain-injured rats was
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

Another allograft model has been used re- evaluated using the Morris Water Maze.
cently to study the ability of the MCV VCP-treated rats were shown to perform sig-
chemokine antagonist MC148 to prolong nificantly better in the Morris Water Maze
graft survival using gene transfer.122 A than the control rats, indicating an increase
heterotropic nonvascular heart transplant, in cognitive recovery.124 The increased spa-
where a neonatal heart is implanted subcuta- tial memory displayed by the VCP-treated
neously, was used in mice. Using plasmid- rats illustrates that its immune modulating
mediated gene transfer, plasmid DNA en- properties are, in fact, beneficial for recov-
coding MC148 was transferred into the donor ery following traumatic brain injury.
heart before transplanting. The presence of
MC148 significantly decreased donor spe-
cific CTL infiltration into graft tissue and D. Spinal Cord Injury Model
For personal use only.

inhibited alloantibody production.


To evaluate the ability of VCP to modu-
late the inflammation and neurological dys-
C. Head Injury Model function following traumatic spinal cord in-
jury, the New York University impact
To evaluate the ability of VCP to modu- contusion model was used. Like with the
late the inflammation and neurological dys- head injury model, adult rats receive a lami-
function following traumatic brain injury, nectomy to expose the dura before being
the lateral fluid percussion brain injury model fastened to the NYU weight-drop impactor
was used. Rats receive a craniotomy to ex- devise. The injury devise causes a very re-
pose the dura before being attached to the producible contusion injury on the spinal
injury devise. The injury devise then sends a cord of the desired severity. Following the
bolus of saline that strikes the surface of the injury, VCP or saline is microinjected into
brain, creating a pressure pulse that causes a the spinal cord parenchyma at multiple bilat-
rapid but temporary displacement of brain eral sites underlying the injury and the wound
tissue. The severity of injury is predeter- is closed. The effect that VCP had on in-
mined and very reproducible. Following the flammatory cell infiltration was then moni-
injury, VCP or saline is microinjected into tored at various post-injury time points by
the cortex underlying the site of injury and hematoxylin and eosin (H&E) staining and
the wound is closed. The effect that VCP the myeloperoxidase (MPO) assay. It was
had on complement activation and inflam- found that VCP-treated spinal cords con-
matory cell infiltration was then monitored tained fewer neutrophils present around the
at various post-injury time points. It was site of the injury.125 To determine whether
found that VCP, when present at the site of the reduction of inflammatory cell infiltra-

175
tion can coincide with functional recovery, causing a large diversity of disease patholo-
injured rats were evaluated using the Basso, gies. The immune response to poxvirus infec-
Beattie, and Bresnahan (BBB) locomotor tion mounted by the host is extremely power-
rating scale, and the grid walking test. VCP- ful and of great complexity. However, the
injected animals performed significantly poxvirus has become an ever more complex
better in the grid walking test and scored and a worthy adversary, evolving new and
higher (although not significantly) on the more powerful immune modulating agents.
BBB locomoter scale for 6 weeks after in- The survival of the virus depends on its abil-
jury. This increase in motor function sug- ity to create and perfect strategies to conceal
gests that rats receiving an injection of VCP itself from this much larger and more sophis-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

at the site of injury exhibit greater neuronal ticated opponent. Presently, the most heavily
sparing and functional recovery. studied aspect of poxvirus infection is the
immune modulating proteins. These amazing
proteins have tremendous potential for use as
E. Antigen-Induced Arthritis (AIA) therapeutic agents for the treatment of many
Model of Chronic Inflammation injuries and diseases, of an inflammatory na-
ture. The creativity and potency of the many
In order to determine the effect that Serp-1 virokines is fascinating and may lend itself to
might have in alleviating chronic inflamma- a new class of therapeutic agents. Someday
tion characteristic of arthritis, an antigen- we may be able to make use of these powerful
induced arthritis (AIA) model was used in proteins that have evolved to near perfection
rabbits. Rabbits were first immunized with for the treatment of diseases that cannot be
For personal use only.

ovalbumin in complete Freund’s adjuvant to treated with conventional therapy. The use of
generate a strong immune response. To in- virokines as immunomodulatory agents is only
duce AIA in a single joint, rabbits received starting to be realized. In addition, new
an intraarticular (i.a.) injection of ovalbu- virokines are constantly being uncovered,
min. Finally, after 2 weeks a second i.a. often with new and novel features. Myxoma
injection of ovalbumin, followed by two daily virus, for example, secretes dozens of pro-
injections of transforming growth factor beta teins from the infected cell, only about five of
(TGF-β) completed the induction of AIA. which have been isolated and characterized to
Serp-1 was then injected i.a. 1 and 2 weeks any extent.
after the final ovalbumin injection, to test its Several animal models have been used
anti-arthritic effects. A histological analysis traditionally to study poxvirus infection, in-
of the synovium later revealed that Serp-1 cluding the CAM, nude mouse, and footpad
injection had a dramatic effect on chronic injection models. In addition, several models
inflammation by reducing inflammatory cell have been developed and employed to evalu-
infiltration, cartilage erosion, and synovium ate the therapeutic effectiveness of various
hyperplasia.126 poxvirus immunomodulatory agents. These
models are designed to closely replicate hu-
man injury or disease and to assess the overall
VII. CONCLUSION usefulness of these poxviral agents. Several
of these proteins have shown great potential
The study of poxvirus infection in vari- in reducing inflammation, thus resulting in a
ous animals is a very exciting and multifac- more favorable outcome. Although the study
eted topic for research. Poxviruses have been of these poxvirus proteins has been quite ex-
found to infect a wide range of animal hosts, tensive, one question that remains is whether

176
they act as virulence factors that promote vi- 2. Howard J., Justus D.E., Totmenin A.V. et
ral replication in host tissue, and if so what al., Molecular mimicry of the inflammation
are the mechanisms, and what do these pro- modulatory proteins (IMPs) of poxviruses:
evasion of the inflammatory response to pre-
teins do in terms of evading host defenses. serve viral habitat, J. Leukoc. Biol., 64, 68–
Until very recently, only a couple of investi- 71, 1998.
gators have attempted to address the role of
3. Moss B., Poxviridae: The Viruses and Their
virokines in whole animals. The data gath-
Replication, in Fundamental Virology, Fields
ered from these experiments could help to B.N., Knipe D.M., Howley P.M., Eds.,
answer many questions, such as why variola Lippincott-Raven, Philadelphia, 1996, 1163–
virus is so deadly even though 80% of its
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

97.
ORFs possess 90% or greater sequence ho-
4. Smith S.A. and Kotwal G.J., Virokines:
mology to that of vaccinia virus. An addi- novel immunomodulatory agents, Exp. Opin.
tional area that needs farther investigation, Biol. Ther., 1, 343–57, 2001.
one of the biggest mysteries in poxvirus re-
5. Buller R.M. and Palumbo G.J., Poxvirus
search is how the viruses acquired these im-
pathogenesis, Microbiol. Rev., 55, 80–122,
mune modulatory proteins. Many of these 1991.
poxvirus proteins possess high sequence ho-
mology to those immune regulatory proteins 6. Fenner F., Mousepox (infectious ectrome-
lia): past, present, and future, Lab Anim Sci.,
found encoded by their natural host. There- 31, 553–9, 1981.
fore, the virus must have stolen them from the
host cell, modified them to be more compact 7. Bhatt P.N. and Jacoby R.O., Effect of vac-
cination on the clinical response, pathogen-
and more powerful, and turned them against
esis and transmission of mousepox, Lab Anim.
For personal use only.

their host. So how did a DNA virus, which Sci., 37, 610–4, 1987.
replicates in the cytoplasm, acquire the nucleic
acid sequence for such proteins? It is widely 8. Kerr P.J. and Best S.M., Myxoma virus in
rabbits, Rev. Sci. Tech., 17, 256–68, 1998.
believed that during coinfection with a
retrovirus, the RNA encoding these proteins 9. Strayer D.S., Skaletsky E., and Sell S., Strain
was converted to DNA and packaged by the differences in Shope fibroma virus, An im-
poxvirus — this hypothesis has yet to be munopathologic study, Am. J. Pathol., 116,
342–58, 1984.
proven. Future experiments will help clear up
these remaining unanswered, often basic, 10. Kurata T., Aoyama Y., and Kitamura T.,
questions about poxvirus infection in animals. Demonstration of vaccinia virus antigen in
brains of postvaccinal encephalitis cases, Jpn.
J. Med. Sci. Biol., 30, 137–47, 1977.

ACKNOWLEDGMENTS 11. Goldstein J.A., Neff J.M., Lane J.M. et al.,


Smallpox vaccination reactions, prophylaxis,
and therapy of complications, Pediatrics, 55,
SAS is a recipient of the Center for Genetics
342–7, 1975.
and Molecular Medicine graduate fellowship. GJK
is currently a senior International Wellcome Trust 12. Antoine G., Scheiflinger F., Dorner F. et
Fellow in biomedical sciences in South Africa. al., The complete genomic sequence of the
modified vaccinia Ankara strain: comparison
with other orthopoxviruses, Virology, 244,
365–96, 1998.
REFERENCES
13. Haig D.M. and Mercer A.A., Ovine dis-
1. Kotwal G.J., Microorganisms and their in- eases Orf, Vet. Res., 29, 311–26, 1998.
teraction with the immune system, J. Leukoc. 14. Yirrell D.L., Reid H.W., Norval M. et al.,
Biol., 62, 415–29, 1997. Immune response of lambs to experimental

177
infection with Orf virus, Vet. Immunol. 26. Amer M., El Gharib I., Rashed A. et al.,
Immunopathol., 22, 321–32, 1989. Human cowpox infection in Sharkia Gover-
norate, Egypt, Int. J. Dermatol., 40, 14–7,
15. Lloyd J.B., Gill H.S., Haig D.M. et al., In
2001.
vivo T-cell subset depletion suggests that
CD4+ T-cells and a humoral immune response 27. Yeruham I., Nir O., Braverman Y. et al.,
are important for the elimination of orf virus Spread of lumpy skin disease in Israeli dairy
from the skin of sheep, Vet. Immunol. herds, Vet. Rec., 137, 91–3, 1995.
Immunopathol., 74, 249–62, 2000.
28. Carn V.M. and Kitching R.P., The clinical
16. Konya J. and Thompson C.H., Molluscum response of cattle experimentally infected with
contagiosum virus: antibody responses in per- lumpy skin disease (Neethling) virus, Arch.
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

sons with clinical lesions and seroepidemiology Virol., 140, 503–13, 1995.
in a representative Australian population, J.
29. Carn V.M. and Kitching R.P., An investi-
Infect. Dis., 179, 701–4, 1999.
gation of possible routes of transmission of
17. Birthistle K. and Carrington D., Mollus- lumpy skin disease virus (Neethling),
cum contagiosum virus, J. Infect., 34, 21–8, Epidemiol. Infect., 114, 219–26, 1995.
1997.
30. Ngichabe C.K., Wamwayi H.M., Barrett
18. Heng M.C., Steuer M.E., Levy A. et al., T. et al., Trial of a capripoxvirus-rinderpest
Lack of host cellular immune response in recombinant vaccine in African cattle,
eruptive molluscum contagiosum, Am. J. Epidemiol. Infect., 118, 63–70, 1997.
Dermatopathol., 11, 248–54, 1989.
31. Chihota C.M., Rennie L.F., Kitching R.P.
19. Charteris D.G., Bonshek R.E., and Tullo et al., Mechanical transmission of lumpy skin
A.B., Ophthalmic molluscum contagiosum: disease virus by Aedes aegypti (Diptera: Cu-
clinical and immunopathological features, Br. licidae), Epidemiol. Infect., 126, 317–21, 2001.
For personal use only.

J. Ophthalmol., 79, 476–81, 1995.


32. Massung R.F., Jayarama V., and Moyer
20. Bugert J.J., Melquiot N., and Kehm R., R.W., DNA sequence analysis of conserved
Molluscum contagiosum virus expresses late and unique regions of swinepox virus: iden-
genes in primary human fibroblasts but does tification of genetic elements supporting phe-
not produce infectious progeny, Virus Genes, notypic observations including a novel G pro-
22, 27–33, 2001. tein-coupled receptor homologue, Virology,
197, 511–28, 1993.
21. Marennikova S.S., Maltseva N.N., Korneeva
V.I. et al., Outbreak of pox disease among 33. Miller R.B. and Olson L.D., Experimental
carnivora (felidae) and edentata, J. Infect. Dis., induction of cutaneous streptococcal abscesses
135, 358–66, 1977. in swine as a sequela to swinepox, Am. J. Vet.
Res., 41, 341–7, 1980.
22. Crouch A.C., Baxby D., McCracken C.M.
et al., Serological evidence for the reservoir 34. Teppema J.S. and De Boer G.F., Ultrastruc-
hosts of cowpox virus in British wildlife, tural aspects of experimental swinepox with
Epidemiol. Infect., 115, 185–91, 1995. special reference to inclusion bodies, Arch.
Virol., 49, 151–63, 1975.
23. Zhukova O.A., Tsanava S.A., and Marennikova
S.S., Experimental infection of domestic cats by 35. Kim U.H., Mukhajonpan V., Nii S. et al.,
cowpox virus, Acta Virol., 36, 329–31, 1992. Ultrastructural study of cell cultures infected
with swinepox and orf viruses, Biken. J., 20,
24. Hinrichs U., van de P.H., and van den Ingh
57–67, 1977.
T.S., Necrotizing pneumonia in a cat caused
by an orthopox virus, J. Comp Pathol., 121, 36. Tripathy D.N. and Hanson L.E., Immunity
191–6, 1999. to fowlpox, Am. J. Vet. Res., 36, 541–4, 1975.
35.
25. Baxby D., Bennett M., and Getty B., Hu-
man cowpox 1969–93: a review based on 54 37. Kent S.J., Zhao A., Best S.J. et al., En-
cases, Br. J. Dermatol., 131, 598–607, 1994. hanced T-cell immunogenicity and protective

178
efficacy of a human immunodeficiency virus and chicken embryos, J. Virol., 57, 670–3,
type 1 vaccine regimen consisting of con- 1986.
secutive priming with DNA and boosting with
48. Miller C.G., Justus D.E., Jayaraman S. et al.,
recombinant fowlpox virus, J. Virol., 72,
Severe and prolonged inflammatory response to
10180–8, 1998.
localized cowpox virus infection in footpads of
38. Leist T.P., Eppler M., and Zinkernagel C5–deficient mice: investigation of the role of
R.M., Enhanced virus replication and inhibi- host complement in poxvirus pathogenesis, Cell
tion of lymphocytic choriomeningitis virus Immunol., 162, 326–32, 1995.
disease in anti-gamma interferon-treated mice,
49. West B.C., Eschete M.L., Cox M.E. et al.,
J. Virol., 63, 2813–9, 1989.
Neutrophil uptake of vaccinia virus in vitro,
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

39. Karupiah G., Fredrickson T.N., Holmes J. Infect. Dis., 156, 597–606, 1987.
K.L. et al., Importance of interferons in re-
50. Jones J.F., Interactions between human neu-
covery from mousepox, J. Virol., 67, 4214–
trophils and vaccinia virus: induction of oxi-
26, 1993.
dative metabolism and virus inactivation,
40. Ramshaw I.A., Ramsay A.J., Karupiah G. Pediatr. Res., 16, 525–9, 1982.
et al., Cytokines and immunity to viral infec-
51. Palumbo G.J., Pickup D.J., Fredrickson
tions, Immunol. Rev., 159, 119–35, 1997.
T.N. et al., Inhibition of an inflammatory
41. Suenaga T., Okuyama T., Yoshida I. et al., response is mediated by a 38–kDa protein of
Effect of Mycobacterium tuberculosis BCG cowpox virus, Virology, 172, 262–73, 1989.
infection on the resistance of mice to ectrome-
52. Tsuru S., Kitani H., Seno M. et al., Mecha-
lia virus infection: participation of interferon
nism of protection during the early phase of a
in enhanced resistance, Infect. Immun., 20,
generalized viral infection. I. Contribution of
312–4, 1978.
phagocytes to protection against ectromelia
For personal use only.

42. Aksenov O.A. and Smorodintsev A.A., virus, J. Gen. Virol., 64, 2021–6, 1983.
Experiences with improvement of the neu-
53. Hansson O., Johansson S.G., and Vahlquist B.,
tralization reaction with vaccinia virus in chick
Vaccinia Gangrenosa with normal humoral anti-
fibroblast cultures, J. Hyg. Epidemiol.
bodies. A case possibly due to deficient cellular
Microbiol. Immunol., 10, 488–98, 1966.
immunity treated with N-methylisatin beta-
43. Leddy J.P., Simons R.L., and Douglas R.G., thiosemicarbazone (Compound 33T57, Marboran),
Effect of selective complement deficiency on Acta Paediatr. Scand., 55, 264–72, 1966.
the rate of neutralization of enveloped viruses
54. McFarland H.F., Pedone C.A., Mingioli
by human sera, J. Immunol., 118, 28–34, 1977.
E.S. et al., The response of human lympho-
44. Hirsch R.L., The complement system: its cyte subpopulations to measles, mumps, and
importance in the host response to viral infec- vaccinia viral antigens, J. Immunol., 125, 221–
tion, Microbiol. Rev., 46, 71–85, 1982. 5, 1980.
45. Brier A.M. and Wohlenberg C., Rosenthal 55. Perrin L.H., Zinkernagel R.M., and
J et al. Inhibition or enhancement of immu- Oldstone M.B., Immune response in humans
nological injury of virus-infected cells, Proc. after vaccination with vaccinia virus: genera-
Natl. Acad. Sci. U.S.A., 68, 3073–7, 1971. tion of a virus-specific cytotoxic activity by
human peripheral lymphocytes, J. Exp. Med.,
46. Wakamiya N., Okada N., Wang Y.L. et al.,
146, 949–69, 1977.
Tumor cells treated with vaccinia virus can
activate the alternative pathway of mouse 56. Moretta L., Moretta A., Hengartner H. et
complement, Jpn. J. Cancer Res., 80, 765– al., On the pathogenesis of perforin defects
70, 1989. and related immunodeficiencies, Immunol.
Today, 21, 593–4, 2000.
47. Ohta H., Yoshikawa Y., Kai C. et al., Effect
of complement depletion by cobra venom 57. Doherty P.C. and Korngold R., Character-
factor on fowlpox virus infection in chickens istics of poxvirus-induced meningitis: virus-

179
specific and non-specific cytotoxic effectors 67. Tewari M., Beidler D.R., and Dixit V.M.,
in the inflammatory exudates, Scand. J. CrmA-inhibitable cleavage of the 70–kDa
Immunol., 18, 1–7, 1983. protein component of the U1 small nuclear
ribonucleoprotein during Fas- and tumor ne-
58. Yang H., Cain C.A., Woan M.C. et al.,
crosis factor- induced apoptosis, J. Biol.
Evaluation of hamster natural cytotoxic cells
Chem., 270, 18738–41, 1995.
and vaccinia-induced cytotoxic cells for Thy-
1.2 homologue by using a mouse monoclonal 68. Kotwal G.J., Poxviral mimicry of comple-
alpha- Thy-1.2 antibody, J. Immunol., 129, ment and chemokine system components:
2239–43, 1982. what’s the end game?, Immunol. Today, 21,
242–8, 2000.
59. Jacoby R.O., Bhatt P.N., and Brownstein
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

D.G., Evidence that NK cells and interferon 69. Sahu A., Isaacs S.N., Soulika A.M. et al.,
are required for genetic resistance to lethal Interaction of vaccinia virus complement con-
infection with ectromelia virus, Arch. Virol., trol protein with human complement proteins:
108, 49–58, 1989. factor I-mediated degradation of C3b to iC3b1
inactivates the alternative complement path-
60. Karupiah G., Coupar B.E., Andrew M.E.
way, J. Immunol., 160, 5596–604, 1998.
et al., Elevated natural killer cell responses in
mice infected with recombinant vaccinia vi- 70. Reynolds D.N., Keeling K.L., Molestina R.
rus encoding murine IL-2, J. Immunol., 144, et al., Heparin binding activity of vaccinia
290–8, 1990. virus complement control protein confers
additional properties of uptake by mast cells
61. Blanden R.V. and Gardner I.D., The cell-
and attatchment to endothelial cells, in: Ad-
mediated immune response to ectromelia vi-
vances in animal virology, Jameel S., Lambris,
rus infection. I. Kinetics and characteristics
J.D., eds., Oxford and IBH, New York, 2000,
of the primary effector T cell response in
337–42.
For personal use only.

vivo, Cell Immunol., 22, 271–82, 1976.


71. Smith S.A., Mullin N.P., Parkinson J. et
62. Andrew M.E., Coupar B.E., and Boyle D.B.,
al., Conserved surface-exposed K/R-X-K/R
Humoral and cell-mediated immune responses
motifs and net positive charge on poxvirus
to recombinant vaccinia viruses in mice,
complement control proteins serve as puta-
Immunol. Cell Biol., 67, 331–7, 1989.
tive heparin binding sites and contribute to
63. Hapel A. and Gardner I., Appearance of inhibition of molecular interactions with hu-
cytotoxic T cells in cerebrospinal fluid of man endothelial cells: a novel mechanism for
mice with ectromelia virus-induced meningi- evasion of host defense, J. Virol., 74, 5659–
tis, Scand. J. Immunol., 3, 311–9, 1974. 66, 2000.
64. Gardner I.D. and Blanden R.V., The cell- 72. Al-Mohanna, Parhar R., and Kotwal G.J.,
mediated immune response to ectromelia vi- Vaccinia virus complement control protein is
rus infection. II. Secondary response in vitro capable of protecting Xenoendothelial cells
and kinetics of memory T cell production in from antibody binding and killing by human
vivo, Cell Immunol., 22, 283–96, 1976. complement and cytotoxic cells, Transplan-
tation, 71, 796–801 2001.
65. Kettle S., Alcami A., Khanna A. et al.,
Vaccinia virus serpin B13R (SPI-2) inhibits 73. Fleming S.B., Haig D.M., Nettleton P. et
interleukin-1beta- converting enzyme and al., Sequence and functional analysis of a
protects virus-infected cells from TNF- and homolog of interleukin-10 encoded by the
Fas- mediated apoptosis, but does not prevent parapoxvirus orf virus, Virus Genes, 21, 85–
IL-1beta-induced fever, J. Gen. Virol., 78, 95, 2000.
677–85, 1997.
74. Xiang Y. and Moss B., IL-18 binding and
66. Nash P., Barrett J., Cao J.X. et al., inhibition of interferon gamma induction by
Immunomodulation by viruses: the myxoma human poxvirus-encoded proteins, Proc. Natl.
virus story, Immunol. Rev., 168, 103–20, 1999. Acad. Sci. U.S.A., 96, 11537–42, 1999.

180
75. Smith V.P., Bryant N.A., and Alcami A., feron receptor homolog M-T7 interacts with
Ectromelia, vaccinia and cowpox viruses en- the heparin-binding domains of chemokines,
code secreted interleukin-18–binding proteins, J. Virol., 71, 4356–63, 1997.
J. Gen. Virol., 81, 1223–30, 2000.
86. Quan L.T., Caputo A., Bleackley R.C.
76. Upton C., Macen J.L., Schreiber M. et al. et al., Granzyme B is inhibited by the
Myxoma virus expresses a secreted protein cowpox virus serpin cytokine response
with homology to the tumor necrosis factor modifier A, J. Biol. Chem., 270, 10377–
receptor gene family that contributes to viral 9, 1995.
virulence, Virology, 184, 370–82, 1991.
87. Turner P.C., Sancho M.C., Thoennes S.R.
77. Loparev V.N., Parsons J.M., Knight J.C. et et al., Myxoma virus Serp2 is a weak in-
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

al., A third distinct tumor necrosis factor re- hibitor of granzyme B and interleukin-
ceptor of orthopoxviruses, Proc. Natl. Acad. 1beta-converting enzyme in vitro and un-
Sci. U.S.A., 95, 3786–91, 1998. like CrmA cannot block apoptosis in cow-
pox virus-infected cells, J. Virol., 73, 6394–
78. Upton C., Mossman K., and McFadden G.,
404, 1999.
Encoding of a homolog of the IFN-gamma
receptor by myxoma virus, Science, 258, 88. Tewari M., Beidler D.R., and Dixit V.M.,
1369–72, 1992. CrmA-inhibitable cleavage of the 70–kDa
protein component of the U1 small nuclear
79. Alcami A. and Smith G.L., A soluble recep-
ribonucleoprotein during Fas- and tumor ne-
tor for interleukin-1 beta encoded by vaccinia
crosis factor-induced apoptosis, J. Biol. Chem.,
virus: a novel mechanism of virus modula-
270, 18738–41, 1995.
tion of the host response to infection, Cell,
71, 153–67, 1992. 89. Upton C., Macen J.L., Wishart D.S. et al.,
Myxoma virus and malignant rabbit fibroma
80. Spriggs M.K., Hruby D.E., Maliszewski
For personal use only.

virus encode a serpin-like protein important


C.R. et al., Vaccinia and cowpox viruses
for virus virulence, Virology, 179, 618–31,
encode a novel secreted interleukin-1– bind-
1990.
ing protein, Cell, 71, 145–52, 1992.
90. Kotwal G.J. and Moss B., Vaccinia virus
81. Vacheron F., Rudent A., Perin S. et al.,
encodes two proteins that are structurally re-
Production of interleukin 1 and tumour ne-
lated to members of the plasma serine pro-
crosis factor activities in bronchoalveolar
tease inhibitor superfamily, J. Virol., 63, 600–
washings following infection of mice by in-
6, 1989.
fluenza virus, J. Gen. Virol., 71, 477–9, 1990.
91. Lomas D.A., Evans D.L., Upton C. et al.,
82. Deane D., McInnes C.J., Percival A. et al.,
Inhibition of plasmin, urokinase, tissue plas-
Orf virus encodes a novel secreted protein
minogen activator, and C1S by a myxoma
inhibitor of granulocyte- macrophage colony-
virus serine proteinase inhibitor, J. Biol.
stimulating factor and interleukin-2, J. Virol.,
Chem., 268, 516–21, 1993.
74, 1313–20, 2000.
83. Senkevich T.G., Bugert J.J., Sisler J.R. et 92. Chernos V.I., Antonova T.P., and Senkevich
al., Genome sequence of a human tumori- T.G., Recombinants between vaccinia and ec-
genic poxvirus: prediction of specific host tromelia viruses bearing the specific pathoge-
response-evasion genes, Science, 273, 813–6, nicity markers of both parents, J. Gen. Virol.,
1996. 66, 621–6, 1985.

84. Graham K.A., Lalani A.S., Macen J.L. et 93. Buller R.M., Chakrabarti S., Moss B. et al.,
al., The T1/35kDa family of poxvirus-secreted Cell proliferative response to vaccinia virus
proteins bind chemokines and modulate leu- is mediated by VGF, Virology, 164, 182–92,
kocyte influx into virus-infected tissues, Vi- 1988.
rology, 229, 12–24, 1997.
94. Fredrickson T.N., Sechler J.M., Palumbo
85. Lalani A.S., Graham K., Mossman K. et G.J. et al., Acute inflammatory response to
al., The purified myxoma virus gamma inter- cowpox virus infection of the chorioallantoic

181
membrane of the chick embryo, Virology, 187, ficient mice by vector-directed IL-2 expres-
693–704, 1992. sion, Nature, 330, 259–62, 1987.
95. Ali A.N., Turner P.C., Brooks M.A. et al., 107. Perera L.P., Goldman C.K., and Waldmann
The SPI-1 gene of rabbitpox virus determines T.A., Comparative assessment of virulence of
host range and is required for hemorrhagic recombinant vaccinia viruses expressing IL-2
pock formation, Virology, 202, 305–14, 1994. and IL-15 in immunodeficient mice, Proc. Natl.
Acad. Sci. U.S.A., 98, 5146–51, 2001.
96. Palumbo G.J., Buller R.M., and Glasgow
W.C., Multigenic evasion of inflammation 108. Hugin A.W., Flexner C., and Moss B., Clear-
by poxviruses, J. Virol., 68, 1737–49, 1994. ance of recombinant vaccinia virus express-
ing IL-2: role of local host immune responses,
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

97. Palumbo G.J., Pickup D.J., Fredrickson


Cell Immunol., 152, 499–509, 1993.
T.N. et al., Inhibition of an inflammatory
response is mediated by a 38–kDa protein of 109. Karupiah G., Blanden R.V., and Ramshaw
cowpox virus, Virology, 172, 262–73, 1989. I.A., Interferon gamma is involved in the re-
covery of athymic nude mice from recombi-
98. Zheng Z.M., Specter S., Zhang J.H. et al.,
nant vaccinia virus/interleukin 2 infection, J.
Further characterization of the biological and
Exp. Med., 172, 1495–503, 1990.
pathogenic properties of erythromelalgia-related
poxviruses, J. Gen. Virol., 73, 2011–9, 1992. 110. Mahalingam S., Farber J.M., and Karupiah
G., The interferon-inducible chemokines
99. Ueda Y., Dumbell K.R., Tsuruhara T. et
MuMig and Crg-2 exhibit antiviral activity In
al., Studies on Cotia virus—an unclassified
vivo, J. Virol., 73, 1479–91, 1999.
poxvirus, J. Gen. Virol., 40, 263–76, 1978.
111. Martinez M.J., Bray M.P., and Huggins
100. Zinkernagel R.M., Althage A., and Jensen
J.W., A mouse model of aerosol-transmitted
F.C., Cell-mediated immune response to lym-
orthopoxviral disease: morphology of ex-
For personal use only.

phocytic choriomeningitis and vaccinia virus


perimental aerosol-transmitted orthopoxviral
in rats, J. Immunol., 119, 1242–7, 1977.
disease in a cowpox virus-BALB/c mouse sys-
101. Kotwal G.J., Miller C.G., and Justus D.E., tem, Arch. Pathol. Lab Med., 124, 362–77, 2000.
The inflammation modulatory protein (IMP)
112. Bray M., Martinez M., Smee D.F. et al.,
of cowpox virus drastically diminishes the
Cidofovir protects mice against lethal aerosol
tissue damage by down-regulating cellular
or intranasal cowpox virus challenge, J. In-
infiltration resulting from complement acti-
fect. Dis., 181, 10–9, 2000.
vation, Mol. Cell Biochem., 185, 39–46, 1998.
113. Smee D.F., Bailey K.W., Wong M. et al.,
102. Soekawa M., Morita C., Moriguchi R. et
Intranasal treatment of cowpox virus respira-
al., Neurovirulence of vaccinia viruses for
tory infections in mice with cidofovir, Antivi-
mice, Zentralbl. Bakteriol., 226, 434–42, 1974.
ral Res., 47, 171–7, 2000.
103. Ginsberg A.H. and Johnson K.P., Vaccinia
114. Smee D.F., Bailey K.W., and Sidwell R.W.,
virus meningitis in mice after intracerebral
Treatment of cowpox virus respiratory infec-
inoculation, Infect. Immun., 13, 1221–7, 1976.
tions in mice with ribavirin as a single agent
104. Buller R.M., Smith G.L., Cremer K. et al., or followed sequentially by cidofovir, Antivir.
Decreased virulence of recombinant vaccinia Chem. Chemother., 11, 303–9, 2000.
virus expression vectors is associated with a
115. Turner G.S., Respiratory infection of mice
thymidine kinase-negative phenotype, Nature,
with vaccinia virus, J. Gen. Virol., 1, 399–
317, 813–5, 1985.
402, 1967.
105. Kotwal G.J., Hugin A.W., and Moss B.,
116. Taylor G., Stott E.J., and Wertz G. et al.,
Mapping and insertional mutagenesis of a
Comparison of the virulence of wild-type thy-
vaccinia virus gene encoding a 13,800–Da
midine kinase (tk)- deficient and tk+ pheno-
secreted protein, Virology, 171, 579–87, 1989.
types of vaccinia virus recombinants after
106. Flexner C., Hugin A., and Moss B., Preven- intranasal inoculation of mice, J. Gen. Virol.,
tion of vaccinia virus infection in immunode- 72, 125–30, 1991.

182
117. Anderson J.B., Smith S.A., Kotwal G.J. et 127. Dahi M.V. and Peterson P.K., Dermato-
al., Vaccinia virus complement control pro- logic manifestations of infectious diseases.
tein inhibits hyperacute xenorejection in a The Upjohn Company, Kalamazoo, 1988,
guinea pig-to-rat heterotopic cervical cardiac 13.
xenograft activation, Transpl. Immunol., in
128. Messud-Petit F., Gelfi J., Delverdier M. et al.,
press, 2002.
Serp2, an inhibitor of the interleukin-1beta-con-
118. Haber E., Can a viral serine proteinase in- verting enzyme, is critical in the pathobiology
hibitor prevent postangioplasty restenosis?, of myxoma virus, J. Virol., 72, 7830–9, 1998.
Circulation, 94, 2694–5, 1996.
129. Chernos V.I., Senkevich T.G., Chelyapov
119. Miller L.W., Dai E., Nash P. et al., Inhibi- N.V. et al., Biologic properties and genome
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

tion of transplant vasculopathy in a rat aortic structure of the recombinants between ec-
allograft model after infusion of anti-inflam- tromelia and rabbitpox viruses, Acta Virol.,
matory viral serpin, Circulation, 101, 1598– 31, 193–202, 1987.
605, 2000.
130. Brandt T.A. and Jacobs B.L., Both carboxy-
120. Lucas A., Liu L., Macen J. et al., Virus- and amino-terminal domains of the vaccinia
encoded serine proteinase inhibitor SERP-1 virus interferon resistance gene, E3L, are re-
inhibits atherosclerotic plaque development quired for pathogenesis in a mouse model, J.
after balloon angioplasty, Circulation, 94, Virol., 75, 850–6, 2001.
2890–900, 1996.
131. Verardi P.H., Jones L.A., Aziz F.H. et al.,
121. Liu L., Lalani A., Dai E. et al., The viral anti- Vaccinia virus vectors with an inactivated
inflammatory chemokine-binding protein M- gamma interferon receptor homolog gene
T7 reduces intimal hyperplasia after vascular (B8R) are attenuated In vivo without a con-
injury, J. Clin. Invest., 105, 1613–21, 2000. comitant reduction in immunogenicity, J.
For personal use only.

Virol., 75, 11–8, 2001.


122. DeBruyne L.A., Li K., Bishop D.K. et al.,
Gene transfer of virally encoded chemokine 132. Zhang W.H., Wilcock D., and Smith G.L.,
antagonists vMIP-II and MC148 prolongs car- Vaccinia virus F12L protein is required for
diac allograft survival and inhibits donor-spe- actin tail formation, normal plaque size, and
cific immunity, Gene Ther., 7, 575–82, 2000. virulence, J. Virol., 74, 11654–62, 2000.
123. Keeling K.L., Hicks R.R., Mahesh J. et al., 133. Price N., Tscharke D.C., Hollinshead M. et
Local neutrophil influx following lateral fluid- al., Vaccinia virus gene B7R encodes an 18–
percussion brain injury in rats is associated kDa protein that is resident in the endoplas-
with accumulation of complement activation mic reticulum and affects virus virulence.
fragments of the third component (C3) of the Virology, 267, 65–79, 2000.
complement system, J. Neuroimmunol., 105,
134. Lin C.L., Chung C.S., Heine H.G. et al.,
20–30, 2000.
Vaccinia virus envelope H3L protein binds to
124. Hicks R.R., Keeling K., Kotwal G.J. et al., cell surface heparan sulfate and is important
Vaccinia virus complement control protein en- for intracellular mature virion morphogenesis
hances functional recovery following traumatic and virus infection in vitro and in vivo., J.
brain injury, J. Neurotrauma, 19, 705–14, 2002. Virol., 74, 3353–65, 2000.
125.Reynolds D.N., Zhang J.-P., Shields C.B. et 135. Tscharke D.C. and Smith G.L., A model
al., Effects of vaccinia virus complement for vaccinia virus pathogenesis and immunity
control protein administration following trau- based on intradermal injection of mouse ear
matic spinal cord injury, Unpublished, 2001. pinnae, J. Gen. Virol., 80, 2751–5, 1999.
126. Maksymowych W.P., Nation N., Nash P. et 136. Wilcock D., Duncan S.A., Traktman P. et
al., Amelioration of antigen induced arthritis al., The vaccinia virus A4OR gene product is
in rabbits treated with a secreted viral serine a nonstructural, type II membrane glycopro-
proteinase inhibitor, J. Rheumatol., 23, 878– tein that is expressed at the cell surface, J.
82, 1996. Gen. Virol., 80, 2137–48, 1999.

183
137. Ruby J., Bluethmann H., and Peschon J.J., 147. Buller R.M., Chakrabarti S., Cooper J.A.,
Antiviral activity of tumor necrosis factor et al., Deletion of the vaccinia virus growth
(TNF) is mediated via p55 and p75 TNF re- factor gene reduces virus virulence, J. Virol.,
ceptors, J. Exp. Med., 186, 1591–6, 1997. 62, 866–74, 1988.
138. Sedger L., Ramshaw I., Condie A. et al., 148. Chernos V.I., Senkevich T.G., Chelyapov
Vaccinia virus replication is independent of N.V. et al., Biologic properties and genome
cellular HSP72 expression which is induced structure of the recombinants between ec-
during virus infection, Virology, 225, 423–7, tromelia and rabbitpox viruses, Acta Virol.,
1996. 31, 193–202, 1987.
139. Rolph M.S., Cowden W.B., Medveczky C.J. 149. Zavagno G., Jaffe B., Esteban M., Role of
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

et al., A recombinant vaccinia virus encoding prostaglandins and non-steroid anti-inflam-


inducible nitric oxide synthase is attenuated matory drugs in the pathogenicity of vaccinia
in vivo, J. Virol., 70, 7678–85, 1996. virus, J. Gen. Virol., 68, 593–600, 1987.
140. Parkinson J.E., Sanderson C.M., and Smith 150. Werner G.T., Jentzsch U., Metzger E. et
G.L., The vaccinia virus A38L gene product al., Studies on poxvirus infections in irradi-
is a 33–kDa integral membrane glycoprotein, ated animals, Arch. Virol., 64, 247–56, 1980.
Virology, 214, 177–88, 1995.
151. Guerin J.L., Gelfi J., Camus C. et al., Char-
141. Martinez-Pomares L., Thompson J.P., and acterization and functional analysis of Serp3:
Moyer R.W., Mapping and investigation of a novel myxoma virus- encoded serpin in-
the role in pathogenesis of the major unique volved in virulence, J. Gen. Virol., 82, 1407–
secreted 35–kDa protein of rabbitpox virus, 17, 2001.
Virology, 206, 591–600, 1995.
152. Robinson A.J., Muller W.J., Braid A.L. et
142. Kettle S., Blake N.W., Law K.M. et al., al., The effect of buprenorphine on the course
For personal use only.

Vaccinia virus serpins B13R (SPI-2) and of disease in laboratory rabbits infected with
B22R (SPI-1) encode M(r) 38.5 and 40K, myxoma virus, Lab Anim., 33, 252–7, 1999.
intracellular polypeptides that do not affect
153. Hnatiuk S., Barry M., Zeng W. et al., Role
virus virulence in a murine intranasal model,
of the C-terminal RDEL motif of the myxoma
Virology, 206, 136–47, 1995.
virus M-T4 protein in terms of apoptosis regu-
143. Tufariello J.M., Cho S., and Horwitz M.S., lation and viral pathogenesis, Virology, 263,
Adenovirus E3 14.7–kilodalton protein, an 290–306, 1999.
antagonist of tumor necrosis factor cytolysis,
154. Lalani A.S., Masters J., Graham K. et al.,
increases the virulence of vaccinia virus in
Role of the myxoma virus soluble CC-
severe combined immunodeficient mice, Proc.
chemokine inhibitor glycoprotein, M- T1,
Natl. Acad. Sci. U.S.A., 91, 10987–91, 1994.
during myxoma virus pathogenesis, Virology,
144. Banham A.H. and Smith G.L., Character- 256, 233–45, 1999.
ization of vaccinia virus gene B12R, J. Gen. 155. Jackson R.J., Hall D.F., and Kerr P.J., Myxoma
Virol., 74, 2807–12, 1993. virus encodes an alpha2,3–sialyltransferase that
enhances virulence, J. Virol., 73, 2376–84, 1999.
145. Thompson J.P., Turner P.C., Ali A.N. et
al., The effects of serpin gene mutations on 156. Messud-Petit F., Gelfi J., Delverdier M. et al.,
the distinctive pathobiology of cowpox and Serp2, an inhibitor of the interleukin-1beta-con-
rabbitpox virus following intranasal inocula- verting enzyme, is critical in the pathobiology
tion of Balb/c mice, Virology, 197, 328–38, of myxoma virus, J. Virol., 72, 7830–9, 1998.
1993.
157. Barry M., Hnatiuk S., Mossman K. et al.,
146. Engelstad M. and Smith G.L., The vaccinia The myxoma virus M-T4 gene encodes a novel
virus 42–kDa envelope protein is required for RDEL-containing protein that is retained
the envelopment and egress of extracellular within the endoplasmic reticulum and is im-
virus and for virus virulence, Virology, 194, portant for the productive infection of lym-
627–37, 1993. phocytes, Virology, 239, 360–77, 1997.

184
158. Mossman K., Lee S.F., Barry M. et al., that interferes with inflammation, Virology,
Disruption of M-T5, a novel myxoma virus 195, 348–63, 1993.
gene member of poxvirus host range super-
161. Opgenorth A., Graham K., Nation N. et al.,
family, results in dramatic attenuation of
Deletion analysis of two tandemly arranged viru-
myxomatosis in infected European rabbits, J.
lence genes in myxoma virus, M11L and myxoma
Virol., 70, 4394–410, 1996.
growth factor, J. Virol., 66, 4720–31, 1992.
159. Mossman K., Nation P., Macen J. et al., 162. Upton C., Macen J.L., Schreiber M. et al.,
Myxoma virus M-T7, a secreted homolog of Myxoma virus expresses a secreted protein
the interferon-gamma receptor, is a critical with homology to the tumor necrosis factor
virulence factor for the development of myx- receptor gene family that contributes to viral
Critical Reviews in Microbiology Downloaded from informahealthcare.com by University Of Alabama Birmingham on 04/29/15

omatosis in European rabbits, Virology, 215, virulence, Virology, 184, 370–82, 1991.
17–30, 1996.
163. Verma P.C. and Rai A., Effects of cyclo-
160. Macen J.L., Upton C., Nation N. et al., phosphamide on pigs infected and vaccinated
SERP1, a serine proteinase inhibitor encoded with swinepox virus, Indian J. Exp. Biol., 27,
by myxoma virus, is a secreted glycoprotein 467–8, 1989.
For personal use only.

185

Vous aimerez peut-être aussi