Skip to main content

The current status and future directions of myxoma virus, a master in immune evasion

Abstract

Myxoma virus (MYXV) gained importance throughout the twentieth century because of the use of the highly virulent Standard Laboratory Strain (SLS) by the Australian government in the attempt to control the feral Australian population of Oryctolagus cuniculus (European rabbit) and the subsequent illegal release of MYXV in Europe. In the European rabbit, MYXV causes a disease with an exceedingly high mortality rate, named myxomatosis, which is passively transmitted by biting arthropod vectors. MYXV still has a great impact on European rabbit populations around the world. In contrast, only a single cutaneous lesion, restricted to the point of inoculation, is seen in its natural long-term host, the South-American Sylvilagus brasiliensis and the North-American S. Bachmani. Apart from being detrimental for European rabbits, however, MYXV has also become of interest in human medicine in the last two decades for two reasons. Firstly, due to the strong immune suppressing effects of certain MYXV proteins, several secreted virus-encoded immunomodulators (e.g. Serp-1) are being developed to treat systemic inflammatory syndromes such as cardiovascular disease in humans. Secondly, due to the inherent ability of MYXV to infect a broad spectrum of human cancer cells, the live virus is also being developed as an oncolytic virotherapeutic to treat human cancer. In this review, an update will be given on the current status of MYXV in rabbits as well as its potential in human medicine in the twenty-first century.

Table of contents

  1. 1.

    The virus

  2. 2.

    History

  3. 3.

    Pathogenesis and disease symptoms

  4. 4.

    Immunomodulatory proteins of MYXV

    1. 4.1.

      MYXV proteins with anti-apoptotic functions

      1. 4.1.1.

        Inhibition of pro-apoptotic molecules

      2. 4.1.2.

        Inhibition by protein-protein interactions by ankyrin repeat viral proteins

      3. 4.1.3.

        Inhibition of apoptosis by enhancing the degradation of cellular proteins

      4. 4.1.4.

        Inhibition of apoptosis by blocking host Protein Kinase R (PKR)

    2. 4.2.

      MYXV proteins interfering with leukocyte chemotaxis

    3. 4.3.

      MYXV serpins that inhibit cellular pro-inflammatory or pro-apoptotic proteases

    4. 4.4.

      MYXV proteins that interfere with leukocyte activation

    5. 4.5.

      MYXV proteins with sequence similarity to HIV proteins

    6. 4.6.

      MYXV proteins with unknown immune function

  5. 5.

    Vaccination strategies against myxomatosis

    1. 5.1.

      Current MYXV vaccines

    2. 5.2.

      Vaccination campaigns to protect European rabbits in the wild

  6. 6.

    Applications of myxoma virus for human medicine

    1. 6.1.

      MYXV proteins as therapeutics for allograft vasculopathy and atherosclerosis

    2. 6.2.

      Applications for MYXV as a live oncolytic virus to treat cancer

  7. 7.

    Discussion and Conclusions

  8. 8.

    List of Abbreviations

References

Author Details

Authors' contributions

Competing interests

Acknowledgements

1. The virus

Myxoma virus (MYXV) is a member of the genus Leporipoxvirus, subfamily Chordopoxvirinae, belonging to the family of Poxviridae. MYXV contains a large linear double stranded deoxyribonucleic acid (DNA) genome consisting of 159 unique viral genes, with diploid terminal inverted repeats (TIRs). Each TIR contains twelve viral genes, nine of which encoding diverse immunomodulating proteins, giving a total of 171 genes [1]. The viral genome is encapsidated within a brick shaped virion, and the complete replication cycle of MYXV takes place exclusively in the cytoplasm of the infected cells from where it expresses a spectrum of host-interactive immunomodulatory proteins [2]. The virus infects only rabbits and European Brown Hares (only occasionally with clinical signs) in the wild, and is nonpathogenic in any tested hosts apart from lagomorphs, but MYXV can replicate in cultured cells from many species, including most human cancer cells which are particularly permissive for MYXV [3, 4].

2. History

The disease caused by MYXV was seen for the first time in laboratory rabbits in 1896 by Giuseppe Sanarelli in Montevideo, Uruguay [4] Sanarelli was invited by the Uruguayan government to found a new Institute of Hygiene in Montevideo that required the production of antibodies, for which European rabbits were necessary. Following their importation from Europe, these rabbits were housed in an outdoor facility that was open to biting arthropod vectors, such as mosquitoes. Shortly thereafter, the European rabbits were struck down by a highly contagious and lethal disease, which was characterized by multiple lesions and tumors of the skin and conjunctiva. Sanarelli named this new rabbit disease myxomatosis and thought it might be caused by an infectious agent, which was confirmed later on by isolation of the causative poxvirus, called MYXV [4].

The European rabbit evolved in Europe and Eastern Asia, and prior to human intervention was not found in the rest of the world, including Australasia and the Americas. During the colonization of new territories, the European rabbit was often taken along as a source of meat or for sport hunting, and this is what occurred during the colonization of Australia in 1860. Between 1860 and 1930, a growing population of feral European rabbits derived from a small number of initial breeding pairs spread over the entire continent and their numbers rose up to an estimated three billion [5]. Since it was estimated that ten rabbits consumed as much vegetation as a single domesticated sheep, the elimination of rabbits was seen as a very necessary step to economically raise the food and wool production of Australia. Rewards were issued to attract new proposals for reducing the size of the feral rabbit population and one of the people who responded was Louis Pasteur, who recommended using Pasteurella cultures that are pathogenic to rabbits. But due to the well-justified skepticism of the Australian government, he was not allowed to release this microbial agent into the wild [4, 6].

Since the original MYXV had a mortality rate of 99.5% in the European rabbit, it was first suggested as a possible means of controlling the Australian feral rabbit population in 1936. Shortly thereafter, the Australian government approved several field release experiments [4, 5]. Before the Second World War, two such field tests were conducted, the first of which took place on an island off the South Australian coast and the second in the dry inland outback region. Both experiments showed almost no prolonged spread of the test virus. After the Second World War, MYXV was again tested in a third region in Southern Australia with more seasonal rainfall [6]. Since it was originally thought that MYXV was mostly spread rabbit-to-rabbit by direct contact, the field-tests were conducted mostly in the dry interior of Australia during autumn, winter and spring but without much success in inducing widespread disease in the rabbit population [5]. However, shortly after these field tests when the rainy season ensued, numerous dead MYXV-infected rabbits were found alongside rivers and streams, infected via the seasonally expanded populations of Culex annulirostris, a mechanical vector for MYXV. However, it rapidly became clear that almost any biting or sucking arthropod could serve as a vector for the virus, which enabled MYXV to spread over large areas [5, 6]. An estimated 400 million rabbits succumbed in as little as one year, and within less than a decade, the feral rabbit population was reduced by 95% [4, 6]. In contrast to South America where the virus co-existed with Sylvilagus sp. hosts in a relatively nonpathogenic relationship, there was no natural host reservoir species for MYXV present in Australia. Therefore, MYXV was constrained to sustain itself in the same host animals that it was killing with such extreme efficiency. This led to a strong selection for less virulent MYXV strains that killed their European rabbit hosts with less efficiency than the originally introduced high pathogenicity strain of MYXV, enabling more extensive replication and spread of the virus by increasing the survival time of its host [4]. At the same time, the selection pressure exerted by this extremely virulent pathogen caused the host rabbit population to select for the genetically resistant strains that were less susceptible to the lethal disease manifestations of myxomatosis. The co-evolutionary selection pressure reduced the overall mortality rate of MYXV in the field rabbits to less than 30% within seven years after introducing MYXV in Australia. After the introduction of MYXV in Great Britain and France, a similar course of co-evolution between MYXV and its rabbit host was seen [7–9] and at present, MYXV is sustained as a chronic enzootic in rabbit populations in America, Southern Europe, New Zealand and Australia [10] (Figure 1).

Figure 1
figure 1

MYXV worldwide distribution. The global distribution of MYXV during the period 01/07/2009-31/12/2009 (copied from [152).

3. Pathogenesis and disease symptoms

The pathogenesis in immunologically naïve European rabbits after infection with MYXV largely depends on the MYXV strain and the rabbit breed or strain. Aside from naturally occurring MYXV strains, frequently used strains in laboratories are the standard laboratory strain (SLS), the Lausanne strain and the Uriarra (Ur) strain. The SLS and the Lausanne strain, which are both virulent MYXV strains originally isolated in South America, were released in Australia and Europe, respectively [4, 7, 11]. The Ur strain was isolated from more resistant field rabbits in Australia in 1953, three years after the first release of SLS. Since the Ur strain had already undergone three years of selection to less virulence, it is often used as a model for attenuated MYXV strains [12]. In this review, the pathogenesis of an SLS infection in laboratory rabbits will be described extensively as the canonical example of myxomatosis, whereas the other pathogenesis patterns will be discussed briefly in comparison to this standard model.

Laboratory rabbits can be infected with MYXV through direct contact with an infected individual, by an arthropod vector, or simply in the laboratory by direct needle inoculation (Figure 2). Through the infection of dendritic cells (DCs) at the primary infection site, and subsequent circulation via the lymphatic and vascular system, MYXV then spreads throughout the body into numerous secondary organs. New myxoma lesions ("myxomes") are formed at multiple secondary sites on the skin where arthropod vectors can acquire high enough titers on their biting organs in order to successfully infect new hosts once they have been serially bitten by the vector (Figure 2) [2, 12, 13].

Figure 2
figure 2

The pathogenesis of nodular myxomatosis. At the site of inoculation (A) viral replication occurs first in the peripheral epidermal cells and then in cells deeper in the dermis (B). This initially induces only a mild primary inflammation and mainly attracts neutrophils at early times. Resident Major Histocompatibility Complex-2 (MHC-2) positive cells, such as Langerhans cells, are also infected (C) [2, 12, 13]. When these cells migrate to the draining lymph node to present viral epitopes in the T-cell zone, they also traffic live virus and T-lymphocytes become infected with MYXV as well (D). This results in wide spread cell death in the T-cell zone of many lymphoid organs and also enables MYXV to spread in a cell-associated manner when these infected T-lymphocytes migrate through the blood and reticuloendothelial circulation. Once in the blood and lymph, MYXV spreads via infected leukocytes to different organs such as the liver, spleen, other distant lymph nodes, testis and epidermis (E). The MYXV replication in these organs induces a strong polymorphonuclear (PMN) cell-influx and in the skin secondary lesions are formed that contain high viral titers, favoring successful spread by mechanical vectors (F) [2, 12].

This course of disease induces a number of diagnostic clinical symptoms that have been well documented for both the SLS and the Lausanne strains in laboratory rabbits, which are representative for the rabbit population used in either the meat industry or as pets. Essentially all infected immunologically naive laboratory rabbits will develop clinical myxomatosis characterized by a pink color and edema at the point of inoculation, followed by progressive conjunctivitis and serous/mucopurulent secretion from eyes and nose [7, 11]. In addition, the infection is accompanied by swelling of the anogenital region and supervening bacterial infections in the respiratory tract as the immune system of the infected host progressively collapses. Animals will usually die 10-14 days post infection (dpi) [14]. Wild Australian rabbits from more resistant populations infected with the SLS strain or laboratory rabbits infected with the Ur strain will display a similar disease course, however usually with a reduced degree of severity, including a delayed onset of clinical symptoms, and a lower overall mortality (Table 1).

Table 1 Comparison of symptoms in wild and domestic rabbits infected with different MYXV strains inducing nodular myxomatosis (adapted from [7, 11]).

Apart from the classical nodular form of myxomatosis discussed above, a second less frequent form of myxomatosis exists. This atypical myxomatosis is believed to spread by direct contact, rather then by biting arthropods [15]. The clinical signs of this second form, named amyxomatous myxomatosis, are predominantly respiratory while skin lesions are few and small [16].

4. Immunomodulatory proteins of MYXV

The reason for the successful replication of MYXV in rabbits is due to the fact that MYXV encodes a vast array of immunomodulatory proteins (Table 2), some of which are rabbit-specific and others are capable of recognizing immune targets from other hosts, including mice or humans. The gene sequences of these proteins are preferentially located at or near the genomic regions closest to the TIRs of the genome [17] and for many of these proteins, their function has already been investigated in vitro and/or in vivo. For other MYXV proteins, however, their functions are solely predicted by comparing them to analogous related genes from eukaryotic cells or other viruses, such as the vaccinia virus (VACV) [18]. In the present review, the functions of these immunomodulatory proteins will be only briefly discussed but for a more elaborate description of the most important examples, we would like to refer to other recent reviews [19–22]. Moreover, aside from this variety of immunomodulatory proteins, MYXV also encodes genes with important functions in disease pathogenenis, e.g. M127L encodes the DNA repair enzyme photolyase which can promote MYXV survival in the environment [23]. As a description of these proteins is beyond the scope of this review, we would also like to refer for these proteins to other reviews [1, 23, 24].

Table 2 Immunomodulatory proteins of MYXV.

4.1. MYXV proteins with anti-apoptotic functions

4.1.1. Inhibition of pro-apoptotic molecules

Multicellular organisms use programmed cell death, also designated apoptosis, as an innate defense mechanism to eliminate cells that pose a threat for the rest of the organism, such as virus-infected cells [25, 26]. Since apoptosis can have a negative effect on viral replication and spread, many viruses, including MYXV, have developed multiple strategies to inhibit this process. One point of interference is the direct inhibition of key pro-apoptotic molecules.

The MYXV protein M-T2 (M002R/L) is a secreted tumor necrosis factor receptor (TNFR) homolog that inhibits apoptosis by binding and subsequently inhibiting rabbit TNF via a conserved ligand-binding domain at the N-terminus of the viral protein [27–29]. A highly conserved viral preligand assembly domain (vPLAD) on the M-T2 protein is responsible for inhibiting intracellular apoptosis, making it the first described viral immunomodulatory protein for which two separate domains with distinct immune inhibiting functions are known [29, 30].

M11L is a viral protein that localizes to the outer mitochondrial membrane through close association with the mitochondrial peripheral benzodiazepine receptor (PBR) component of the permeabilitytransition (PT) pore. There, M11L prevents the release of mitochondrial cytochrome c induced by staurosporine or protoporphyrin IX (PPIX), which are ligands of PBR [31, 32]. Furthermore, M11L is also able to inhibit apoptosis by binding with B-cell lymphoma 2 (BCL2)-antagonist/killer (Bak), which, following pro-apoptotic stimuli, is also able to release cytochrome c and subsequently induce cell death [26, 33, 34]. Another function for M11L is the ability to interact with activated Bcl-2 associated × protein (Bax), which it inhibits when it is translocated from the cytoplasm to the outer mitochondrial membrane during viral infection [35].

Although not yet demonstrated experimentally, M146R is also a potential inhibitor of the pro-apoptotic Bcl-2 family, in the sense that the MYXV protein is closely related to the VACV protein N1L, which has been shown to bind and inhibit Bcl-2 proteins [36, 37]. Recently, it was shown that N1L also retains a role in influencing the Toll-like receptor (TLR) signaling pathway [38]. Also, and even more recently, it was found that the VACV ortholog N1L of ectromelia virus (ECTV), the causative agent of mousepox, interferes with host T cell function in vivo, independently of TLR signaling. This was also illustrated by the impairment of in vivo ECTV spread when N1L was deleted [39]. This study provided different results for the virulence factor N1L than those previously predicted in vitro, emphasizing again the importance of studying virulence factors in a natural virus-host system in vivo.

4.1.2. Inhibition by protein-protein interactions by ankyrin repeat viral proteins

Several MYXV proteins contain ankyrin (ANK) repeats and a "Pox protein Repeats of Ankyrin C-terminal" (PRANC) domain (an F-box-like domain), which enable the binding with unique cellular protein targets [40, 41]. Through these protein-protein interactions MYXV ANK-repeat proteins act as substrate adaptors that can recruit cellular proteins for ubiquitination and degradation via the 26S proteasome. This is the case for M-T5 (M005R/L), which binds via an adaptor protein called Skp1 with the cellular protein cullin-1 in vitro [42]. Cullin-1 is an E3 ubiquitin ligase, which normally binds to cyclin-dependent kinase (CDK) inhibitors, enzymes that are otherwise degraded in a cell cycle fashion by proteasomes. The binding of M-T5 with Skp1/cullin-1 augments the proteosomal degradation of CDK inhibitors, like p27-Kip, thereby stimulating the progression of the cell cycle past the G0/G1 checkpoint that could otherwise trigger an apoptosis response when the cell cycle stalls at this checkpoint [42, 43]. In vivo experiments have shown that M-T5 is necessary for MYXV to spread secondary immune organs in the infected rabbit host [44], suggesting that M-T5 permits virus spread via infected lymphocytes that traffic to multiple distal immune organs [20].

The myxoma nuclear factor (MNF) M150R is crucial for productive viral infection in vivo and is predicted to interfere with apoptosis by inhibiting Nucleus Factor-κB (NF-κB). This prediction is based on the analogy with ANK-repeats and the nuclear location of M150R when compared to cellular NF-κB inhibitors (IκBs) [45]. NF-κB is an important transcription factor that is activated by pro-inflammatory stimuli, subsequently translocating from the cytoplasm to the nucleus where it upregulates host genes critical for immunity, inflammation and apoptosis [46]. IκBs inhibit NF-κB by interfering with the translocation of the active transcription factor dimer to the nucleus and hence, a similar action is predicted for M150R [45, 47]. More recently, an alternate possibility of inhibition has been proposed, where M150R directs the ubiquitin ligase complex to new substrate targets, such as NF-κB, for ubiquitination and subsequently degradation [48]. Both M148R and M149R are also members of the MYXV ANK-repeat family and although the virulence of infection with M148R and M149R deletion mutants was reduced in in vivo experiments, no specific function has yet been determined for these proteins to date [49].

4.1.3. Inhibition of apoptosis by enhancing the degradation of cellular proteins

Apoptosis can also be inhibited by downregulating immune-receptors located on the cell surface, thereby decreasing the chance of recognition by apoptosis-inducing immune cells such as cytotoxic T-lymphocytes [50]. M153R, also known as Myxoma Virus-Leukemia-Associated Protein (MV-LAP), is a ubiquitin ligase which achieves the downregulation of a number of immune-receptors at the cell surface, such as major histocompatibility complex 1 (MHC-1), cluster of differentiation (CD)-4 and Fas (CD95), by augmented endocytosis or by decreased recycling to the surface [13, 50, 51]. For Fas, this downregulation was also accomplished by degradation [50]. The MYXV protein M143 is also a ubiquitin ligase, but it remains to be determined whether this viral protein displays a similar anti-apoptotic role as MV-LAP [1].

M-T4 (M004R/L) is an anti-apoptotic intracellular virulence factor that has been suggested to function within the endoplasmic reticulum (ER) of infected cells where it binds to calreticulin [13, 52, 53]. Its anti-apoptotic action, necessary for in vivo spreading throughout the infected rabbit host, is still not understood but it might be explained by a possible interference with the function of BAP31, an ER-localized host protein involved in (i) the egress of MHC-1 and other proteins, (ii) inhibiting apoptosis and (iii) downregulating MHC-1 expression respectively [13, 54].

4.1.4. Inhibition of apoptosis by blocking host Protein Kinase R (PKR)

Protein kinase R (PKR) is upregulated by interferon (IFN) and is catalytically activated by double stranded RNA (dsRNA), which can be produced during replication of many classes of viruses, including poxviruses. The activation of PKR causes many downstream anti-viral effects, including apoptosis of the cell, thereby impeding viral spread, and thus many viruses have evolved anti-PKR countermeasures [55]. For example, the VACV K3L protein binds PKR, hereby inhibiting the phosphorylation of the eukaryotic initiation factor 2 alpha (eIF2α), an important regulator of translation initiation [56]. Interestingly, the gene sequence of the protein M156R of MYXV shows extensive sequence similarities with VACV K3L and a similar function of M156R was confirmed since M156R protein was shown to be an efficient substrate for PKR in vitro [57]. Indeed, MYXV shows a partial resistance against type-1 IFN in human cells in vitro [58], indicating that MYXV is capable of interfering with IFN signaling in virus-infected cells, even though it is relatively inhibited when the anti-viral state is pre-induced prior to virus infection [59]. It has been shown that MYXV can partially inhibit type-1 IFN signaling by blocking the interferon-induced activation of the Janus kinase Tyk2 in MYXV-infected human cells. The responsible MYXV protein however, is yet to be determined [58]. A conserved region of M029L, located in its C-terminal domain sequence, shows strong resemblance with the dsRNA binding region of the VACV gene E3L, which is responsible decreasing the activation of PKR [1, 56, 60].

In addition, M062R, M063R and M064R are all orthologs of the VACV host range protein C7L, although only M062R shows similar host range capabilities as C7L as determined by gene swap experiments in the context of a VACV infection [1, 61]. M063R shows sequence similarity with its C-terminal half a with a glutamate-rich domain of the cellular death-domain associated protein, Daxx, leading to the hypothesis that M063R can inhibit Fas-associated cell death through interfering with the function of Daxx [1, 61, 62] However, this model remains unproven and it is still not completely understood under which circumstances Daxx functions as either a pro-apoptotic or an anti-apoptotic protein [63, 64]. Alternatively, it is hypothesized that M063R functions as a host range gene like M062R, but that it exerts this function through a different pathway [62, 65]. Recent evidence suggests that M062R, but not C7L, binds to a specific host anti-viral protein called "sterile alpha motif domain containing protein 9" (SAMD9) implicated in human inflammatory disorders, suggesting that even these closely related viral host range factors may block cellular self-defense pathways by different mechanisms [65].

4.2. MYXV proteins interfering with leukocyte chemotaxis

The coordinated recruitment of leukocytes at the site of infection is an important aspect of the early immune response against viruses [66]. Therefore, it is not surprising that poxviruses such as MYXV have also developed multiple ways to interfere with this process. M-T1 (M001R/L) is a secreted viral protein that is unnecessary for productive viral replication but it can bind and inhibit chemokines of the CC-subfamily in vitro, thereby inhibiting the directional migration and activation of these cells in vivo [66, 67]. In contrast, M-T1 was not able to bind or inhibit chemokines of the CXC subfamily, at least as assessed by chemical crosslinking in vitro, such as CXCL8 (ie IL-8, in humans), and this could explain why neutrophils are the dominant early responder leukocytes at sites of infection in experimentally MYXV-infected laboratory rabbits [12, 67].

M104L is a small hydrophobic MYXV protein which shares amino acid identity of 42% over a 40-residue region (containing a receptor domain implicated in chemokine receptor signaling) with a subset of the transmembrane domains of the IL-8 receptor analogue ORF74 of Ateline herpesvirus 3, which is believed to inhibit the signaling of a yet to be determined chemokine receptor via heterodimerization, hereby preventing the formation of functional receptor dimers [1]. Another viral MYXV protein, M-T7 (M007R/L), which is an abundantly secreted 37 kDa glycoprotein from MYXV-infected cells, binds rabbit interferon-γ (IFN-γ), hereby inhibiting interferon-γ (IFN-γ) through competitive binding [68]. In addition, it was shown that M-T7 protein is able to bind chemokines of the CXC, CC and C subfamilies in vitro in a pan-species specific fashion, thereby blocking the chemokine-mediated recruiting of leukocytes from many species, including rabbits, mice and humans [69].

4.3. MYXV serpins that inhibit cellular pro-inflammatory or pro-apoptotic proteases

Serine protease inhibitors (serpins) are the largest family of cellular protease inhibitors and MYXV encodes three members, each with its own specific functions [1]. Serp-1 (M008.1R/L) is a secreted MYXV serpin that is able to bind and inhibit the functions of several human cellular proteases, such as plasmin, urokinase, plasminogene activator and C1s of the complement cascade [70]. However, more research is needed to clarify the interactions of Serp-1 with similar cellular proteases in the rabbit and the implications of such interactions on the course of disease. Serp-2 (M151R), on the other hand, is an intracellular cysteine proteinase that decreases the secretion of bioactive IL-1β by inhibiting the IL-1β-converting enzyme (ICE) in vitro [71]. Serp-2 is also a weak inhibitor of granzyme B, which plays an important role during T cell-induced apoptosis [72, 73]. Serp-3 (M152R) is the third MYXV protein with a canonical predicted serpin structure, although it has several apparent domain deletions compared to the gene sequences of other serpins [74]. When this Serp-3 gene is deleted from MYXV, no secondary skin lesions are observed in rabbits in vivo, despite the fact that no decrease in viral replication nor an increase of apoptosis can be detected in cultured cells [74].

4.4. MYXV proteins that interfere with leukocyte activation

MYXV encodes M141R, a structural homologue of CD200, which is a cellular membrane-bound immune cell surface protein that is present on a broad range of cells [1, 75]. Cellular CD200 functions as a ligand for a receptor (CD200R) which is only present on myeloid cells and is thought to function by transmitting inhibitory signals [75]. It is believed that M141R (also called vCD200 or vOX-2) sends inhibitory signals to CD200R+ DCs or macrophages, hereby decreasing the capacity of antigen presenting cells to activate lymphocytes [76, 77]. This theory is supported by some in vivo data, where an increased activation of circulating lymphocytes and macrophages was observed when rabbits were infected with a recombinant MYXV mutant lacking M141R [76].

Two other predicted MYXV proteins, M121R and M122R, have been proposed to bind with MHC-1 on the cell surface, since they have a similar gene sequence as two natural ligands of MHC-I, namely the natural killer cell group 2 (NKG2) in humans and the lymphocyte 49 (LY-49) receptors in mice [1]. M128L (vCD47) is another MYXV immunomodulator, encoding a membrane-associated protein that resembles the cellular immune CD47, also known as the integrin associated protein (IAP) [1]. CD47 functions as a stimulatory cell surface ligand for the signal regulatory protein α (SIRPα) on the surface of myeloid cells [78]. CD47, together with SIRPα, form a cell-cell communication system mediating immune cellular functions such as adhesion, mobility, activation and phagocytosis of leukocytes [78, 79]. The current hypothesis is that M128L competes with CD47 for binding to SIRPα, hereby disrupting the leukocyte cell functions described above [80].

Pro-inflammatory caspases also play an important role in the immune responses by regulating the activation and secretion of various pro-inflammatory cytokines like IL-1β and IL-18 [81]. These caspases are activated by a multiprotein complex named the inflammasome [82]. M13L is a pyrin domain (PYD)-containing MYXV protein that binds with a pyrin-containing component (called ASC) of the inflammasome, thereby inhibiting the activation of IL-1β and IL-18 [81]. In addition, M13L also directly inhibits cellular NF-κB signaling by binding NF-κB1/p105 protein, which regulates the secretion of pro-inflammatory cytokines such as TNF, IL-6 and monocyte chemotactic protein (MCP)-1, thereby giving M13L a dual immune-subversive role by inhibiting both the inflammasome and NF-κB signaling [83]. Finally, M154R encodes a protein presenting 50% identity with M2L, a vaccinia virus gene that was also shown to inhibit induction of NF-κB activation [45, 84].

4.5. MYXV proteins with sequence similarity to HIV proteins

Most genes of MYXV have orthologous family members in other poxviruses, with the exception of M129R and M130R. These two MYXV proteins exhibit a partial sequence similarity with key proteins from another highly immunomodulatory virus, namely human immunodeficiency virus (HIV). The MYXV protein M129R partly resembles the V3 loop of the HIV glycoprotein 120 (gp120) (critical for HIV binding with CCR5), but lacks a transmembrane domain and a signal sequence. The gp120 protein is involved in HIV entry through binding with CD4 and CCR5 or CXCR4. These receptors normally fulfill important roles in T-lymphocyte effector functions [85]. It has been hypothesized that M129R may fulfill a similar role as gp120, namely mediating viral entry, but due to the lack of a transmembrane domain, it is not clear yet if M129R is expressed at all on the viral envelope of MYXV [60]. The MYXV protein M130R is expressed as a late protein and exhibits some sequence similarity with the glutamine rich region of the HIV protein transactivator (Tat) [60, 86]. Tat is partially released from HIV-infected cells, although other analyses also revealed a nuclear location for this protein [87]. Many different functions have been accredited to Tat, including RNA binding, inhibition of T-cell proliferation, induction of apoptosis in T-cells and neurons, inhibition of phagocytosis, decrease of apoptosis in infected cells, interfering with NK-cells, etc. Because of the large variety in functions of Tat, the MYXV protein M130R needs to be tested for all these functions to elucidate if M130R actually shares any functional relationship with HIV Tat [60, 88].

4.6. MYXV proteins with unknown immune function

Finally, there are also still many MYXV proteins for which no function or sequence similarity with other obvious immunomodulatory counterpart proteins have been found to date. The myxoma growth factor (MGF), also designated M010L, is a secreted virulence factor with similarity to transforming growth factor-alpha and epidermal growth factor (including six conserved cysteine residues involved in Epidermal growth factor (EGF) receptor binding), which is necessary for a successful infection in vivo, although the underlying mechanism is still to be determined [89]. Because of a strong sequence similarity with the VACV protein B19R from strain Copenhagen (called B18R in the VACV strain Western Reserve), it was originally thought that M135R was a viral scavenger of type-1 IFN, thereby inhibiting this ligand family [60, 90–92]. However, this hypothesis has not been validated since all binding and inhibition assays conducted to date, designed to demonstrate that M135R can indeed interact with type-1 IFN, were negative [90]. Nevertheless, M135R remains an important virulence factor for myxomatosis, so more research on the exact function of this protein is warranted.

Finally, the MYXV protein M144R shows similarity to the VACV proteins C3L (31% by means of blast search) and B5R (37% by means of alignment analysis) [1, 60]. These latter proteins have a complement-binding and structural function respectively, making it tempting to speculate that M144R also possesses these functions, although this still needs to be confirmed [60].

5. Vaccination strategies against myxomatosis

5.1. Current MYXV vaccines

Prevention of myxomatosis is a matter of great importance for industrial/domestic rabbit farms for laboratory and pet rabbits because of its high prevalence and mortality rate [10]. This prevention can be partly achieved through controlling vectors such as mosquitoes and fleas. However, such sanitary measures are rarely sufficient and therefore, an effective vaccination strategy is desired for successful protection against MYXV [93]. Since a robust cellular immunity is necessary for protection against MYXV, inactivated vaccines have generally proven unsuccessful [93]. Plasmid-based subunit vaccines were also unable to protect rabbits from disease, even though both antigen-specific cell-mediated and humoral immune responses were induced [94]. Therefore, only live vaccines, which can be divided into two groups, are commercially available to date. The first group of vaccines consists of heterologous vaccines based on the closely related but nonpathogenic leporipoxvirus Shope fibroma virus (SFV), due to the close antigenic resemblance of this virus with MYXV [93]. SFV originates from the Eastern cottontail rabbit (Sylvilagus floridanus) and provides considerable cross-protection against MYXV without causing disease in European rabbits older than two weeks [2, 10]. Possible reasons for this attenuated phenotype of SFV might lie in the partial or complete deletion of several SFV genes found in MYXV, such as those encoding predicted (eg M150R, M139R) or confirmed (eg Serp-1, M135R) virulence factors [95]. Current commercial SFV vaccines are based on the original Shope OA strain, Boerlage strain or closely related strains [93]. The main disadvantage of these vaccines is that they only induce a limited amount of protective immunity that lasts no longer than three months [96, 97]. Contradictory findings have been reported as to whether SFV-based vaccines can completely prevent clinical signs of myxomatosis upon infection [98, 99]. A possible explanation in this regard could be differences in the adjuvant used or the dosage of vaccine virus used [98]. In addition to adjuvant and dosage of the vaccine virus used, also the vaccination schemes (route, number of injections) could influence the level of protection [99] The second group of vaccines consists of homologous attenuated live vaccines, such as the SG33, Borghi or BTK/RB/84 strains of MYXV [93, 100]. SG33 is a homologous vaccine strain, derived from the Lausanne strain of MYXV by serial passages on RK13 rabbit kidney and chicken embryo cells at 33°C [101], during which genomic deletions were introduced [15]. Specifically, a large deletion near the right end of the genome was seen during preliminary analysis [102]. This was later confirmed when the SG33 genome was analyzed [15, 45]. These deletions include the loss of important immunomodulatory genes, such as Serp-2, M148R, M149R, M150R, M152R, M153R, M154R and M-T1 [15, 45]. In addition, a mutation of the M143 gene was reported in the SG33 strain [15]. The recent analysis of the entire SG33 genome also gave new information concerning the origin of this vaccine strain. It was established that its present sequence composition is the result of field recombination between a wild-type Lausanne strain and a Californian MSD-derived vaccine strain [45]. The Borghi strain is a homologous vaccine strain, derived from the Californian strain MSD of MYXV by serial passages in RK13 cells. The attenuation of the Borghi strain could be due to truncation of Serp-2, M-T4 and/or a mutation in the M121 gene [15]. These homologous MYXV-based vaccines induce a stronger protective immune response than any of the current SFV vaccines and they induce disease protection at least four months after vaccination [93]. The main disadvantage of these vaccines, however, is a reported immunodepression in young rabbits after vaccination. Still, no problems were reported after vaccination with SG33 of young Angora rabbits, which is a breed known to be particularly sensitive to the myxomatosis virus under field conditions [103]. Other side effects reported include lesions of the skin, edema and rash at the injection point, as well as secondary myxoma lesions after vaccination with SG33 [103]. Recently, vaccination with an Uriarra strain deficient for M063R was also evaluated giving long-term results similar to those of heterologous live vaccines [104]. More potent vaccine candidates have been constructed through deletion of one or more virulence genes (e.g. M-T7 or M007L/R, M010L and M011L) in the naturally attenuated MYXV Ur strain [7, 12, 105]. Vaccination with these vaccines, however, is accompanied by mild clinical symptoms in adult rabbits, thereby making them still too virulent for widespread use as a vaccine. When all three deletions were created simultaneously in the same vaccine, no symptoms were observed in test rabbits but the long-term protection against wild-type MYXV was also limited [7, 12, 105]. By inserting a VP60 (rabbit hemorrhagic disease (RHD) capsid component that induces a protective antibody response against RHD [106]) construct in the SG33 strain, another potent vaccine strain was created which protected rabbits against both MYXV and RHD [107]. Despite showing promising preliminary results, this recombinant vaccine is still not commercially available [93].

In general, the currently recommended vaccination scheme against myxomatosis consists of a primary vaccination with SFV at three or four weeks of age and a booster vaccination with a homologous MYXV-based vaccine three weeks to two months later, depending if the rabbits are exposed to a high or low infection risk. The rabbits then need to be re-vaccinated every four to six months, depending on the infection risk [93].

5.2. Vaccination campaigns to protect European rabbits in the wild

The European rabbit is also an important part of Europe's ecosystems and it is an essential source of food for at least 29 predatory species [108]. MYXV, together with RHD and hunting, is an important reason for the poor condition of some of these indigenous prey populations, such as the Iberian lynx and many raptors [109]. Management strategies such as captivity breeding, restocking, translocations, predator control or habitat management, aimed at raising the number of rabbits for hunting and conservation purposes, have been attempted but they have had very little impact on the overall rabbit population numbers, especially in Mediterranean ecosystems [110]. Therefore, management strategies should include a vaccination program, with adequate procedures [97, 110]. However, all of the currently available commercial vaccines are not suitable for vaccination in the wild since they have to be administered individually [93, 111]. Therefore, attempts have been made to develop transmissible vaccines that are able to serially spread throughout the rabbit populations in the wild. For this reason, twenty MYXV field strains were isolated from the wild and were evaluated for their virulence and horizontal transmissibility [112]. One of these field strains, isolate 6918, showed appropriate characteristics for safety and immunization, which also remained the case after inserting a VP60 construct for inducing additional immunization against RHD [112–114]. The attenuated phenotype of isolate 6918, in comparison to the Lausanne MYXV strain from which it originated, is possibly caused by frameshift mutations that severely disrupted at least four different viral genes (M009L, M036L, M135R, M148R) [115]. The disruption of M135R and M148R could be important since they are known virulence factors [49, 90, 115]. The use of such a vaccine, with a successful horizontal transmission capacity, could eventually lead to the protection of a sufficient portion of the rabbit population after capturing, directly vaccinating and releasing a small number of rabbits [112]. Therefore, a field test was authorized on a small island near mainland Europe with a dense rabbit population. After inoculating one fourth of the population, a limited transmission was seen with a seroconversion in 50% of the non-inoculated rabbits [116]. Although promising, the relatively low transmissibility might be an obstacle for immunization in less densely populated areas, so more field tests are needed to fully investigate the potential of this vaccine [117].

Apart from the vaccination itself, the time of vaccination has to be taken into account also in order to maximize the vaccination campaign's effectiveness in areas where myxomatosis is already enzootic. The time of vaccination would be most efficient before the start of the yearly epidemics, since the herd immunity will then be at its lowest and numerous non-immune kits will have been born [117]. Research has shown that there is a two-month window between the birth of susceptible animals and the appearance of a myxomatosis epidemic, which would be the best time for introducing a new MYXV strain in an already endemic area [118]. This, however, could be difficult to implement since the timing of the epidemics varies yearly [117].

6. Applications of myxoma virus for human medicine

6.1. MYXV proteins as therapeutics for allograft vasculopathy and atherosclerosis (Table 3)

Table 3 Clinically adapted MYXV proteins for treating inflammatory diseases.

Transplant vasculopathy is a pathological syndrome that is associated with chronic rejection of organ transplants and it is the leading cause of transplanted organ loss after the first year post-transplant [119]. Its histopathology is characterized by narrowing of the vascular lumen as a result of proliferation of smooth muscle cells and fibroblasts. Furthermore, material of the extracellular matrix is deposited and activated leukocytes infiltrate the vessel wall. There is also a large histopathological resemblance between graft vasculopathy and non-transplant forms of atherosclerosis, due to overlapping pathogenetic pathways [120]. Atherosclerosis can be caused by many factors, including hyperlipidemia, diabetes and hypertension [121]. Numerous proteases are important factors for the development of atherosclerosis, since they help regulate atherosclerosis-related vascular remodeling. An important group of these proteases are the serine proteases since they not only mediate matrix breakdown, but also activate and release diverse pro-inflammatory cytokines and growth factors, thus activating a wide spectrum of leukocyte receptors and promoting increased leukocyte migration into damaged tissues [122]. The ability of purified recombinant Serp-1 protein to inhibit several of these serine protease in a species-independent manner raises the possibility that this viral protein could attenuate the vascular inflammatory responses in a wide variety of cardiovascular diseases, such as atherosclerosis, transplantation rejection and injury vasculopathy [122, 123]. Indeed, there are potentially numerous applications in human medicine, for example treating other diseases driven by systemic inflammation. Recently, successful human Phase II clinical trials have been reported using purified Serp-1 protein to treat patients with acute coronary syndromes [124, 125].

The MYXV protein Serp-2, on the other hand, is an intracellular regulator that selectively binds and inhibits mediators of T-cell killing, enabling a generalized reduction of arterial inflammation through a granzyme B/perforin dependent pathway. This may prolong certain specific T-cell functions, allowing subsets of T-cells to provide anti-inflammatory actions or alternatively it may initiate apoptotic responses in disease-causing macrophages and/or smooth muscle cells [122]. Serp-2 protein has not yet been tested in clinical trials [19].

Another important feature of vascular inflammation is the attraction of leukocytes by chemokines to specific sites in the vasculature and their migration through surrounding tissues [121]. The M-T1 and M-T7 secreted proteins of MYXV, which inhibit in a species-independent fashion a variety of diverse chemokines, block the chemotaxis of leukocytes into these sites of inflammation, subsequently reducing the allograft rejection in a murine aortic allograft model [126–129]. In the case of immune-based joint diseases like rheumatoid arthritis, high quantities of pro-inflammatory cytokines, such as TNF, are found in the synovial fluids and the amount of TNF elevation closely correlates with disease severity. Subsequently, TNF is considered an important target for treatment [129]. The MYXV protein M-T2 has high affinity for rabbit TNF, and also can bind and inhibit human TNF-receptors, but more research is still needed to determine if this MYXV protein would have therapeutic potential [28–30, 129].

6.2. Applications for MYXV as a live oncolytic virus to treat cancer

MYXV is also considered a promising oncolytic virus that is essentially harmless to non-lagomorphs, including mice and humans, but nevertheless is able to selectively infect and kill many diverse human tumor cells. This translational application of MYXV is based on the genetic alterations in intracellular signaling pathways commonly found in cancer cells, which allows the transformed cells to continue dividing without being controlled by normal cellular checkpoints [20, 130–136]. On the other hand, these cellular checkpoints also form an important part of the innate self-protection mechanisms against viral infection, and therefore, most transformed cells tend to be more susceptible to many viral infections [134].

The absence of such viral protection mechanisms in cancer cells makes the selective replication possible for at least some oncolytic viruses that normally cannot propagate successfully in human tissues, such as MYXV [134]. More specifically for MYXV, malignant cancer cells in general lose the ability to induce the TNF/IFN-β synergistic anti-viral state, a feature that largely determines the host range of MYXV in primary human cells [130]. In addition, the host range of MYXV in human cancer cells is also influenced by oncogenic activation and tumor suppressor dysfunction [137]. Indeed, MYXV has been shown to be capable of infecting a wide variety of human cancer cell lines in vitro and it has been proven to be a safe and potent oncolytic virus in vivo [132, 138, 139]. However, in order to be suitable for anti-tumoral therapy in the clinic, MYXV also needs to cause regression of metastatic tumors in immune competent patients [132]. When administered systemically, MYXV was able to decrease murine B16 melanoma lung metastasis in an immune competent mouse model. This regression of tumors was significantly enhanced by the chemotherapeutic rapamycin [140], an effect which was later on shown to be achieved by reconfiguring the internal cell signaling environment of many cancer cells to one which is more optimal for productive virus replication [134]. A more elaborate description of MYXV as an oncolytic virus has recently been reviewed by Liu et al. [19], and the particular exploitation of MYXV to purge cancer cells from human bone marrow transplants is described by Rahman et al. [141].

7. Discussion and conclusions

Research on MYXV has been conducted for many years and the interest in this virus is basically two-fold. On the one hand, MYXV is an important pathogen for the European rabbit population and due to economical reasons and ecological consequences, MYXV merits attention for both population control strategies in places (like Australia) where feral rabbit populations have become damaging, as well as vaccination strategies for situations (like commercial rabbit breading operations, or for native rabbits in Europe) where myxomatosis is a detrimental disease. On the other hand, MYXV is also gaining more interest from a therapeutic point of view now that it has been shown to have great potential for treatment of certain inflammatory-based diseases and cancer in humans. Thus, it is of utmost importance to continue to acquire detailed knowledge on the behavior and pathogenesis of this rabbit-specific viral pathogen in both permissive (ie rabbit) and nonpermissive (especially human) hosts.

It has become clear over the years that developing effective vaccines and efficient vaccination strategies for rabbits are not as straightforward as originally thought. The major problems encountered are balancing the need to induce a potent acquired immune response without being accompanied by disease symptoms. Deletions of MYXV virulence genes, but maintaining virus replication fitness, is probably the best approach for a longer lasting immunity, as has been shown by Adams et al. [105]. However, improved potency in terms of the desired immune response was often accompanied by the appearance of some clinical symptoms in vaccinated rabbits, indicating that the level of protection that can be achieved to date with the available vaccines on the market might remain the best approach since these vaccines are of no risk for the rabbit itself.

Another possibility for creating a more potent immune reaction against MYXV could be insertion of important immunogenic genes of MYXV into heterologous vaccine platforms. Such approach has already been reported for canine influenza, equine influenza virus, West Nile virus and bovine viral diarrhea virus, where virulence genes of these viruses were inserted into an equine herpesvirus type 1 (EHV1) vaccine [142–145]. The rationale behind the insertion of MYXV immunogenic genes into an equine pathogen is based on the fact that EHV1 grows extremely well in rabbit cells and hence, rabbit kidney (RK13) are the most commonly used in vitro cell line to grow EHV1 [146, 147]. Still, despite this efficient growth in vitro, it needs to be determined whether EHV1 is capable of inducing robust immune responses in rabbits in vivo. Preliminary experiments, however, have shown that rabbits are susceptible for EHV1 infection (Van de Walle et al., unpublished results), indicating that a recombinant EHV1 vector expressing MYXV proteins could prove a novel strategy to efficiently protect rabbits against MYXV. In addition, another possible viral vector vehicle for immunization against MYXV could be exploited with nonpathogenic variants of Vesicular Stomatitis Virus (VSV) since it has shown excellent replication kinetics in the rabbit during experiments for vaccine development [148–150].

Numerous discoveries have been made concerning the immunomodulatory capabilities of MYXV since the sequencing of the full MYXV genome, although the functions of many candidate proteins remain undetermined to date. For some of the latter class of proteins, a putative function had been sometimes ascribed based on sequence similarity with other immunomodulatory viral or cellular proteins, but these conjectures have not always been validated, such as the cases of M063 [1, 62] and M135 [90]. This indicates that one needs to be judicious when predicting how a viral protein functions solely based on sequence or structural similarities. Rather, a true biological role can only be assigned after thorough in vitro and in vivo experimentations.

In terms of clinical applications, several of the MYXV immunomodulatory proteins have become promising candidates for treatment of inflammation-based disorders. To date, the first of these virus-derived drugs (Serp-1) has behaved particularly well in humans, with essentially no clinical adverse side effects, in Phase I and II clinical trials. MYXV as an oncolytic virus candidate also shows considerable potential, both against murine cancers in immunocompetent mouse hosts, as well as human cancers xenografted into immunodeficient mice. MYXV appears to be capable of substantially counteracting malignancies derived from many tissues, especially in combination with rapamycin, and the live virus has exhibited essentially no detectable side effects in all murine hosts tested, even mice that are extensively immunocompromised. In the future, insertion of selective anti-cancer therapeutic genes [131] and/or deletion of MYXV virulence genes [151] could further augment its safety against all vertebrate hosts (including rabbits) and therefore improve its clinical suitability. For example, the Interleukin-15 (IL-15) gene has been inserted into the MYXV genome and this recombinant virus remained capable of replicating in vitro to a degree resembling wild MYXV [131]. Very promising results, concerning safety improvement, have also been obtained by deleting the host range genes M063 or M135 since either deletion showed significant attenuation in its natural rabbit host while retaining and even improving the viral oncolytic capabilities compared to wild MYXV [151]. The exploitation of MYXV constructs that are nonpathogenic in all known vertebrate hosts, but still potently oncolytic against human cancers, will hopefully make MYXV an important factor in improving the outcome of treatment for numerous cancers in the future [19].

Abbreviations

(ANK):

ankyrin

(BCL2):

B-cell lymphoma 2

(Bak):

(BCL2)-antagonist/killer

(Bax):

Bcl-2 associated × protein

(CD):

cluster of differentiation

(CDK):

cyclin-dependent kinase

(DCs):

dendritic cells

(DNA):

double stranded deoxyribonucleic acid

(dsRNA):

double stranded RNA

(ECTV):

ectromelia virus

(ER):

endoplasmic reticulum

(EGF):

epidermal growth factor

(eIF2α):

eukaryotic initiation factor 2 alpha

(gp120):

glycoprotein 120

(ICE):

IL-1β-converting enzyme

(IAP):

integrin associated protein

(IFN):

interferon

(IFN-γ):

interferon-γ

(IL-15):

Interleukin-15

(Ur):

Lausanne strain and the Uriarra

(LY-49):

lymphocyte 49

(MHC-1):

major histocompatibility complex 1

(MCP)-1:

monocyte chemotactic protein

(MGF):

myxoma growth factor

(MNF):

myxoma nuclear factor

(MYXV):

Myxoma virus

(MV-LAP):

Myxoma Virus-Leukemia-Associated Protein

(NKG2):

natural killer cell group 2

(IκBs):

NF-κB inhibitors

(NF-κB):

Nucleus Factor-Κb

(PBR):

peripheral benzodiazepine receptor

(PT):

permeabilitytransition

(PRANC):

Pox protein Repeats of Ankyrin C-terminal

(PKR):

Protein Kinase R

(PPIX):

protoporphyrin IX

(PYD):

pyrin domain

(RHD):

rabbit hemorrhagic disease

(RK):

rabbit kidney

(serpins):

Serine protease inhibitors

(SFV):

Shope fibroma virus

(SIRPα):

signal regulatory protein α

(SLS):

Standard Laboratory Strain

(SAMD9):

sterile alpha motif domain containing protein 9

(TIRs):

terminal inverted repeats

(TLR):

Toll-like receptor

(Tat):

transactivator

(TNFR):

tumor necrosis factor receptor

(VACV):

vaccinia virus

(VSV):

Vesicular Stomatitis Virus

(vPLAD):

viral preligand assembly domain.

References

  1. Cameron C, Hota-Mitchell S, Chen L, Barrett J, Cao JX, Macaulay C, Willer D, Evans D, McFadden G: The complete DNA sequence of myxoma virus. Virology. 1999, 264: 298-318. 10.1006/viro.1999.0001.

    CAS  PubMed  Google Scholar 

  2. Kerr P, McFadden G: Immune responses to myxoma virus. Viral Immunol. 2002, 15: 229-246. 10.1089/08828240260066198.

    CAS  PubMed  Google Scholar 

  3. Saari SA, Rudback E, Niskanen M, Syrjala P, Nylund M, Anttila M: Contagious mucocutaneous dermatitis of the mountain hare (Lepus timidus): pathology and cause. J Wildl Dis. 2005, 41: 775-782.

    PubMed  Google Scholar 

  4. Fenner F, Ratcliffe F: Myxomatosis. 1965, University Press, Cambridge Eng

    Google Scholar 

  5. Fenner F: Deliberate introduction of the European rabbit, Oryctolagus cuniculus, into Australia. Rev Sci Tech. 2010, 29: 103-111.

    CAS  PubMed  Google Scholar 

  6. Burnet FM: Myxomatosis as a method of biological control against the Australian rabbit. Am J Public Health Nations Health. 1952, 42: 1522-1526. 10.2105/AJPH.42.12.1522.

    PubMed Central  CAS  PubMed  Google Scholar 

  7. Best SM, Kerr PJ: Coevolution of host and virus: the pathogenesis of virulent and attenuated strains of myxoma virus in resistant and susceptible European rabbits. Virology. 2000, 267: 36-48. 10.1006/viro.1999.0104.

    CAS  PubMed  Google Scholar 

  8. Ross J, Tittensor AM: The establishment and spread of myxomatosis and its effect on rabbit populations. Philos Trans R Soc Lond B Biol Sci. 1986, 314: 599-606. 10.1098/rstb.1986.0075.

    CAS  PubMed  Google Scholar 

  9. Bartrip PW: Myxomatosis in 1950s Britain. 20 Century Br Hist. 2008, 19: 83-105.

    CAS  PubMed  Google Scholar 

  10. Fenner F, Fantini B: Biological Control of vertebrate pests: the history of myxomatosis - an experiment in evolution. 1999, CABI Publishing, Oxford

    Google Scholar 

  11. Jeklova E, Leva L, Matiasovic J, Kovarcik K, Kudlackova H, Nevorankova Z, Psikal I, Faldyna M: Characterisation of immunosuppression in rabbits after infection with myxoma virus. Vet Microbiol. 2008, 129: 117-130. 10.1016/j.vetmic.2007.11.039.

    CAS  PubMed  Google Scholar 

  12. Best SM, Collins SV, Kerr PJ: Coevolution of host and virus: cellular localization of virus in myxoma virus infection of resistant and susceptible European rabbits. Virology. 2000, 277: 76-91. 10.1006/viro.2000.0505.

    CAS  PubMed  Google Scholar 

  13. Zuniga MC: Lessons in detente or know thy host: the immunomodulatory gene products of myxoma virus. J Biosci. 2003, 28: 273-285. 10.1007/BF02970147.

    CAS  PubMed  Google Scholar 

  14. Robinson AJ, Muller WJ, Braid AL, Kerr PJ: The effect of buprenorphine on the course of disease in laboratory rabbits infected with myxoma virus. Lab Anim. 1999, 33: 252-257. 10.1258/002367799780578129.

    CAS  PubMed  Google Scholar 

  15. Cavadini P, Botti G, Barbieri I, Lavazza A, Capucci L: Molecular characterization of SG33 and Borghi vaccines used against myxomatosis. Vaccine. 2010, 28: 5414-5420. 10.1016/j.vaccine.2010.06.017.

    CAS  PubMed  Google Scholar 

  16. Marlier D, Cassart D, Boucraut-Baralon C, Coignoul F, Vindevogel H: Experimental infection of specific pathogen-free New Zealand White rabbits with five strains of amyxomatous myxoma virus. J Comp Pathol. 1999, 121: 369-384. 10.1053/jcpa.1999.0335.

    CAS  PubMed  Google Scholar 

  17. Nash P, Barrett J, Cao JX, Hota-Mitchell S, Lalani AS, Everett H, Xu XM, Robichaud J, Hnatiuk S, Ainslie C, Seet BT, McFadden G: Immunomodulation by viruses: the myxoma virus story. Immunol Rev. 1999, 168: 103-120. 10.1111/j.1600-065X.1999.tb01286.x.

    CAS  PubMed  Google Scholar 

  18. Goebel SJ, Johnson GP, Perkus ME, Davis SW, Winslow JP, Paoletti E: The complete DNA sequence of vaccinia virus. Virology. 1990, 179: 247-266, 517-563. 10.1016/0042-6822(90)90294-2.

    CAS  PubMed  Google Scholar 

  19. Liu J, Wennier S, McFadden G: The immunoregulatory properties of oncolytic myxoma virus and their implications in therapeutics. Microbes Infect. 2010, 12: 1144-1152. 10.1016/j.micinf.2010.08.012.

    PubMed Central  CAS  PubMed  Google Scholar 

  20. Stanford MM, Werden SJ, McFadden G: Myxoma virus in the European rabbit: interactions between the virus and its susceptible host. Vet Res. 2007, 38: 299-318. 10.1051/vetres:2006054.

    CAS  PubMed  Google Scholar 

  21. Seet BT, Johnston JB, Brunetti CR, Barrett JW, Everett H, Cameron C, Sypula J, Nazarian SH, Lucas A, McFadden G: Poxviruses and immune evasion. Annu Rev Immunol. 2003, 21: 377-423. 10.1146/annurev.immunol.21.120601.141049.

    CAS  PubMed  Google Scholar 

  22. Johnston JB, Wang G, Barrett JW, Nazarian SH, Colwill K, Moran M, McFadden G: Myxoma virus M-T5 protects infected cells from the stress of cell cycle arrest through its interaction with host cell cullin-1. J Virol. 2005, 79: 10750-10763. 10.1128/JVI.79.16.10750-10763.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  23. Bennett CJ, Webb M, Willer DO, Evans DH: Genetic and phylogenetic characterization of the type II cyclobutane pyrimidine dimer photolyases encoded by Leporipoxviruses. Virology. 2003, 315: 10-19. 10.1016/S0042-6822(03)00512-9.

    CAS  PubMed  Google Scholar 

  24. Webb MAM: An investigation of factors affecting poxvirus genetic stability. 2004, National Library of Canada, Ottawa, Canada

    Google Scholar 

  25. Hengartner MO: The biochemistry of apoptosis. Nature. 2000, 407: 770-776. 10.1038/35037710.

    CAS  PubMed  Google Scholar 

  26. Taylor JM, Barry M: Near death experiences: poxvirus regulation of apoptotic death. Virology. 2006, 344: 139-150. 10.1016/j.virol.2005.09.032.

    CAS  PubMed  Google Scholar 

  27. Schreiber M, McFadden G: The myxoma virus TNF-receptor homologue (T2) inhibits tumor necrosis factor-alpha in a species-specific fashion. Virology. 1994, 204: 692-705. 10.1006/viro.1994.1585.

    CAS  PubMed  Google Scholar 

  28. Schreiber M, Rajarathnam K, McFadden G: Myxoma virus T2 protein, a tumor necrosis factor (TNF) receptor homolog, is secreted as a monomer and dimer that each bind rabbit TNFalpha, but the dimer is a more potent TNF inhibitor. J Biol Chem. 1996, 271: 13333-13341. 10.1074/jbc.271.23.13333.

    CAS  PubMed  Google Scholar 

  29. Sedger LM, Osvath SR, Xu XM, Li G, Chan FK, Barrett JW, McFadden G: Poxvirus tumor necrosis factor receptor (TNFR)-like T2 proteins contain a conserved preligand assembly domain that inhibits cellular TNFR1-induced cell death. J Virol. 2006, 80: 9300-9309. 10.1128/JVI.02449-05.

    PubMed Central  CAS  PubMed  Google Scholar 

  30. Schreiber M, Sedger L, McFadden G: Distinct domains of M-T2, the myxoma virus tumor necrosis factor (TNF) receptor homolog, mediate extracellular TNF binding and intracellular apoptosis inhibition. J Virol. 1997, 71: 2171-2181.

    PubMed Central  CAS  PubMed  Google Scholar 

  31. Castedo M, Perfettini JL, Kroemer G: Mitochondrial apoptosis and the peripheral benzodiazepine receptor: a novel target for viral and pharmacological manipulation. J Exp Med. 2002, 196: 1121-1125. 10.1084/jem.20021758.

    PubMed Central  CAS  PubMed  Google Scholar 

  32. Everett H, Barry M, Sun X, Lee SF, Frantz C, Berthiaume LG, McFadden G, Bleackley RC: The myxoma poxvirus protein, M11L, prevents apoptosis by direct interaction with the mitochondrial permeability transition pore. J Exp Med. 2002, 196: 1127-1139. 10.1084/jem.20011247.

    PubMed Central  CAS  PubMed  Google Scholar 

  33. Everett H, Barry M, Lee SF, Sun X, Graham K, Stone J, Bleackley RC, McFadden G: M11L: a novel mitochondria-localized protein of myxoma virus that blocks apoptosis of infected leukocytes. J Exp Med. 2000, 191: 1487-1498. 10.1084/jem.191.9.1487.

    PubMed Central  CAS  PubMed  Google Scholar 

  34. Wang G, Barrett JW, Nazarian SH, Everett H, Gao X, Bleackley C, Colwill K, Moran MF, McFadden G: Myxoma virus M11L prevents apoptosis through constitutive interaction with Bak. J Virol. 2004, 78: 7097-7111. 10.1128/JVI.78.13.7097-7111.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  35. Su J, Wang G, Barrett JW, Irvine TS, Gao X, McFadden G: Myxoma virus M11L blocks apoptosis through inhibition of conformational activation of Bax at the mitochondria. J Virol. 2006, 80: 1140-1151. 10.1128/JVI.80.3.1140-1151.2006.

    PubMed Central  CAS  PubMed  Google Scholar 

  36. Aoyagi M, Zhai D, Jin C, Aleshin AE, Stec B, Reed JC, Liddington RC: Vaccinia virus N1L protein resembles a B cell lymphoma-2 (Bcl-2) family protein. Protein Sci. 2007, 16: 118-124.

    PubMed Central  CAS  PubMed  Google Scholar 

  37. Cooray S, Bahar MW, Abrescia NG, McVey CE, Bartlett NW, Chen RA, Stuart DI, Grimes JM, Smith GL: Functional and structural studies of the vaccinia virus virulence factor N1 reveal a Bcl-2-like anti-apoptotic protein. J Gen Virol. 2007, 88: 1656-1666. 10.1099/vir.0.82772-0.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Gonzalez JM, Esteban M: A poxvirus Bcl-2-like gene family involved in regulation of host immune response: sequence similarity and evolutionary history. Virol J. 2010, 7: 59-10.1186/1743-422X-7-59.

    PubMed Central  PubMed  Google Scholar 

  39. Gratz MS, Suezer Y, Kremer M, Volz A, Majzoub M, Hanschmann KM, Kalinke U, Schwantes A, Sutter G: N1L is an ectromelia virus virulence factor and essential for in vivo spread upon respiratory infection. J Virol. 2011, 85: 3557-3569. 10.1128/JVI.01191-10.

    PubMed Central  CAS  PubMed  Google Scholar 

  40. Mercer AA, Fleming SB, Ueda N: F-box-like domains are present in most poxvirus ankyrin repeat proteins. Virus Genes. 2005, 31: 127-133. 10.1007/s11262-005-1784-z.

    CAS  PubMed  Google Scholar 

  41. Sedgwick SG, Smerdon SJ: The ankyrin repeat: a diversity of interactions on a common structural framework. Trends Biochem Sci. 1999, 24: 311-316. 10.1016/S0968-0004(99)01426-7.

    CAS  PubMed  Google Scholar 

  42. Werden SJ, Lanchbury J, Shattuck D, Neff C, Dufford M, McFadden G: The myxoma virus m-t5 ankyrin repeat host range protein is a novel adaptor that coordinately links the cellular signaling pathways mediated by Akt and Skp1 in virus-infected cells. J Virol. 2009, 83: 12068-12083. 10.1128/JVI.00963-09.

    PubMed Central  CAS  PubMed  Google Scholar 

  43. Johnston JB, Wang G, Barrett JW, Nazarian SH, Colwill K, Moran M, McFadden G: Myxoma virus M-T5 protects infected cells from the stress of cell cycle arrest through its interaction with host cell cullin-1. J Virol. 2005, 79: 10750-10763. 10.1128/JVI.79.16.10750-10763.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  44. Mossman K, Lee SF, Barry M, Boshkov L, McFadden G: Disruption of M-T5, a novel myxoma virus gene member of poxvirus host range superfamily, results in dramatic attenuation of myxomatosis in infected European rabbits. J Virol. 1996, 70: 4394-4410.

    PubMed Central  CAS  PubMed  Google Scholar 

  45. Camus-Bouclainville C, Fiette L, Bouchiha S, Pignolet B, Counor D, Filipe C, Gelfi J, Messud-Petit F: A virulence factor of myxoma virus colocalizes with NF-kappaB in the nucleus and interferes with inflammation. J Virol. 2004, 78: 2510-2516. 10.1128/JVI.78.5.2510-2516.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  46. Chen ZJ: Ubiquitin signalling in the NF-kappaB pathway. Nat Cell Biol. 2005, 7: 758-765. 10.1038/ncb0805-758.

    PubMed Central  CAS  PubMed  Google Scholar 

  47. Baeuerle PA: IkappaB-NF-kappaB structures: at the interface of inflammation control. Cell. 1998, 95: 729-731. 10.1016/S0092-8674(00)81694-3.

    CAS  PubMed  Google Scholar 

  48. Blanie S, Gelfi J, Bertagnoli S, Camus-Bouclainville C: MNF, an ankyrin repeat protein of myxoma virus, is part of a native cellular SCF complex during viral infection. Virol J. 2010, 7: 56-10.1186/1743-422X-7-56.

    PubMed Central  PubMed  Google Scholar 

  49. Blanie S, Mortier J, Delverdier M, Bertagnoli S, Camus-Bouclainville C: M148R and M149R are two virulence factors for myxoma virus pathogenesis in the European rabbit. Vet Res. 2009, 40: 11-10.1051/vetres:2008049.

    PubMed Central  PubMed  Google Scholar 

  50. Guerin JL, Gelfi J, Boullier S, Delverdier M, Bellanger FA, Bertagnoli S, Drexler I, Sutter G, Messud-Petit F: Myxoma virus leukemia-associated protein is responsible for major histocompatibility complex class I and Fas-CD95 down-regulation and defines scrapins, a new group of surface cellular receptor abductor proteins. J Virol. 2002, 76: 2912-2923. 10.1128/JVI.76.6.2912-2923.2002.

    PubMed Central  CAS  PubMed  Google Scholar 

  51. Mansouri M, Bartee E, Gouveia K, Hovey Nerenberg BT, Barrett J, Thomas L, Thomas G, McFadden G, Fruh K: The PHD/LAP-domain protein M153R of myxomavirus is a ubiquitin ligase that induces the rapid internalization and lysosomal destruction of CD4. J Virol. 2003, 77: 1427-1440. 10.1128/JVI.77.2.1427-1440.2003.

    PubMed Central  CAS  PubMed  Google Scholar 

  52. Barry M, Hnatiuk S, Mossman K, Lee SF, Boshkov L, McFadden G: The myxoma virus M-T4 gene encodes a novel RDEL-containing protein that is retained within the endoplasmic reticulum and is important for the productive infection of lymphocytes. Virology. 1997, 239: 360-377. 10.1006/viro.1997.8894.

    CAS  PubMed  Google Scholar 

  53. Hnatiuk S, Barry M, Zeng W, Liu L, Lucas A, Percy D, McFadden G: Role of the C-terminal RDEL motif of the myxoma virus M-T4 protein in terms of apoptosis regulation and viral pathogenesis. Virology. 1999, 263: 290-306. 10.1006/viro.1999.9946.

    CAS  PubMed  Google Scholar 

  54. Zuniga MC: A pox on thee! Manipulation of the host immune system by myxoma virus and implications for viral-host co-adaptation. Virus Res. 2002, 88: 17-33. 10.1016/S0168-1702(02)00118-1.

    CAS  PubMed  Google Scholar 

  55. Sadler AJ, Williams BR: Interferon-inducible antiviral effectors. Nat Rev Immunol. 2008, 8: 559-568. 10.1038/nri2314.

    PubMed Central  CAS  PubMed  Google Scholar 

  56. Langland JO, Jacobs BL: The role of the PKR-inhibitory genes, E3L and K3L, in determining vaccinia virus host range. Virology. 2002, 299: 133-141. 10.1006/viro.2002.1479.

    CAS  PubMed  Google Scholar 

  57. Ramelot TA, Cort JR, Yee AA, Liu F, Goshe MB, Edwards AM, Smith RD, Arrowsmith CH, Dever TE, Kennedy MA: Myxoma virus immunomodulatory protein M156R is a structural mimic of eukaryotic translation initiation factor eIF2alpha. J Mol Biol. 2002, 322: 943-954. 10.1016/S0022-2836(02)00858-6.

    CAS  PubMed  Google Scholar 

  58. Wang F, Barrett JW, Shao Q, Gao X, Dekaban GA, McFadden G: Myxoma virus selectively disrupts type I interferon signaling in primary human fibroblasts by blocking the activation of the Janus kinase Tyk2. Virology. 2009, 387: 136-146. 10.1016/j.virol.2009.02.013.

    CAS  PubMed  Google Scholar 

  59. Bartee E, Mohamed MR, Lopez MC, Baker HV, McFadden G: The addition of tumor necrosis factor plus beta interferon induces a novel synergistic antiviral state against poxviruses in primary human fibroblasts. J Virol. 2009, 83: 498-511. 10.1128/JVI.01376-08.

    PubMed Central  CAS  PubMed  Google Scholar 

  60. Barrett JW, Cao JX, Hota-Mitchell S, McFadden G: Immunomodulatory proteins of myxoma virus. Semin Immunol. 2001, 13: 73-84. 10.1006/smim.2000.0298.

    CAS  PubMed  Google Scholar 

  61. Meng X, Chao J, Xiang Y: Identification from diverse mammalian poxviruses of host-range regulatory genes functioning equivalently to vaccinia virus C7L. Virology. 2008, 372: 372-383. 10.1016/j.virol.2007.10.023.

    PubMed Central  CAS  PubMed  Google Scholar 

  62. Barrett JW, Shun Chang C, Wang G, Werden SJ, Shao Z, Barrett C, Gao X, Belsito TA, Villenevue D, McFadden G: Myxoma virus M063R is a host range gene essential for virus replication in rabbit cells. Virology. 2007, 361: 123-132. 10.1016/j.virol.2006.11.015.

    CAS  PubMed  Google Scholar 

  63. Curtin JF, Cotter TG: Live and let die: regulatory mechanisms in Fas-mediated apoptosis. Cell Signal. 2003, 15: 983-992. 10.1016/S0898-6568(03)00093-7.

    CAS  PubMed  Google Scholar 

  64. Salomoni P, Khelifi AF: Daxx: death or survival protein?. Trends Cell Biol. 2006, 16: 97-104. 10.1016/j.tcb.2005.12.002.

    CAS  PubMed  Google Scholar 

  65. Liu J, Wennier S, Zhang L, McFadden G: M062 is a host range factor essential for myxoma virus pathogenesis and functions as an antagonist of host SAMD9 in human cells. J Virol. 2011, 85: 3270-3282. 10.1128/JVI.02243-10.

    PubMed Central  CAS  PubMed  Google Scholar 

  66. Lalani AS, Masters J, Graham K, Liu L, Lucas A, McFadden G: Role of the myxoma virus soluble CC-chemokine inhibitor glycoprotein, M-T1, during myxoma virus pathogenesis. Virology. 1999, 256: 233-245. 10.1006/viro.1999.9617.

    CAS  PubMed  Google Scholar 

  67. Lalani AS, Ness TL, Singh R, Harrison JK, Seet BT, Kelvin DJ, McFadden G, Moyer RW: Functional comparisons among members of the poxvirus T1/35 kDa family of soluble CC-chemokine inhibitor glycoproteins. Virology. 1998, 250: 173-184. 10.1006/viro.1998.9340.

    CAS  PubMed  Google Scholar 

  68. Upton C, Mossman K, McFadden G: Encoding of a homolog of the IFN-gamma receptor by myxoma virus. Science. 1992, 258: 1369-1372. 10.1126/science.1455233.

    CAS  PubMed  Google Scholar 

  69. Boomker JM, Luttikhuizen DT, Veninga H, de Leij LF, The TH, de Haan A, van Luyn MJ, Harmsen MC: The modulation of angiogenesis in the foreign body response by the poxviral protein M-T7. Biomaterials. 2005, 26: 4874-4881. 10.1016/j.biomaterials.2004.11.059.

    CAS  PubMed  Google Scholar 

  70. Macen JL, Upton C, Nation N, McFadden G: SERP1, a serine proteinase inhibitor encoded by myxoma virus, is a secreted glycoprotein that interferes with inflammation. Virology. 1993, 195: 348-363. 10.1006/viro.1993.1385.

    CAS  PubMed  Google Scholar 

  71. Petit F, Bertagnoli S, Gelfi J, Fassy F, Boucraut-Baralon C, Milon A: Characterization of a myxoma virus-encoded serpin-like protein with activity against interleukin-1 beta-converting enzyme. J Virol. 1996, 70: 5860-5866.

    PubMed Central  CAS  PubMed  Google Scholar 

  72. Chavez-Galan L, Arenas-Del Angel MC, Zenteno E, Chavez R, Lascurain R: Cell death mechanisms induced by cytotoxic lymphocytes. Cell Mol Immunol. 2009, 6: 15-25. 10.1038/cmi.2009.3.

    PubMed Central  CAS  PubMed  Google Scholar 

  73. Turner PC, Sancho MC, Thoennes SR, Caputo A, Bleackley RC, Moyer RW: Myxoma virus Serp2 is a weak inhibitor of granzyme B and interleukin-1beta-converting enzyme in vitro and unlike CrmA cannot block apoptosis in cowpox virus-infected cells. J Virol. 1999, 73: 6394-6404.

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Guerin JL, Gelfi J, Camus C, Delverdier M, Whisstock JC, Amardeihl MF, Py R, Bertagnoli S, Messud-Petit F: Characterization and functional analysis of Serp3: a novel myxoma virus-encoded serpin involved in virulence. J Gen Virol. 2001, 82: 1407-1417.

    CAS  PubMed  Google Scholar 

  75. Barclay AN, Wright GJ, Brooke G, Brown MH: CD200 and membrane protein interactions in the control of myeloid cells. Trends Immunol. 2002, 23: 285-290. 10.1016/S1471-4906(02)02223-8.

    CAS  PubMed  Google Scholar 

  76. Cameron CM, Barrett JW, Liu L, Lucas AR, McFadden G: Myxoma virus M141R expresses a viral CD200 (vOX-2) that is responsible for down-regulation of macrophage and T-cell activation in vivo. J Virol. 2005, 79: 6052-6067. 10.1128/JVI.79.10.6052-6067.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Zhang L, Stanford M, Liu J, Barrett C, Jiang L, Barclay AN, McFadden G: Inhibition of macrophage activation by the myxoma virus M141 protein (vCD200). J Virol. 2009, 83: 9602-9607. 10.1128/JVI.01078-09.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. van Beek EM, Cochrane F, Barclay AN, van den Berg TK: Signal regulatory proteins in the immune system. J Immunol. 2005, 175: 7781-7787.

    CAS  PubMed  Google Scholar 

  79. Matozaki T, Murata Y, Okazawa H, Ohnishi H: Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009, 19: 72-80. 10.1016/j.tcb.2008.12.001.

    CAS  PubMed  Google Scholar 

  80. Cameron CM, Barrett JW, Mann M, Lucas A, McFadden G: Myxoma virus M128L is expressed as a cell surface CD47-like virulence factor that contributes to the downregulation of macrophage activation in vivo. Virology. 2005, 337: 55-67. 10.1016/j.virol.2005.03.037.

    CAS  PubMed  Google Scholar 

  81. Johnston JB, Barrett JW, Nazarian SH, Goodwin M, Ricciuto D, Wang G, McFadden G: A poxvirus-encoded pyrin domain protein interacts with ASC-1 to inhibit host inflammatory and apoptotic responses to infection. Immunity. 2005, 23: 587-598. 10.1016/j.immuni.2005.10.003.

    CAS  PubMed  Google Scholar 

  82. Petrilli V, Papin S, Tschopp J: The inflammasome. Curr Biol. 2005, 15: R581-10.1016/j.cub.2005.07.049.

    CAS  PubMed  Google Scholar 

  83. Rahman MM, Mohamed MR, Kim M, Smallwood S, McFadden G: Co-regulation of NF-kappaB and inflammasome-mediated inflammatory responses by myxoma virus pyrin domain-containing protein M013. PLoS Pathog. 2009, 5: e1000635-10.1371/journal.ppat.1000635.

    PubMed Central  PubMed  Google Scholar 

  84. Gedey R, Jin XL, Hinthong O, Shisler JL: Poxviral regulation of the host NF-kappaB response: the vaccinia virus M2L protein inhibits induction of NF-kappaB activation via an ERK2 pathway in virus-infected human embryonic kidney cells. J Virol. 2006, 80: 8676-8685. 10.1128/JVI.00935-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  85. Qin S, Rottman JB, Myers P, Kassam N, Weinblatt M, Loetscher M, Koch AE, Moser B, Mackay CR: The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. J Clin Invest. 1998, 101: 746-754. 10.1172/JCI1422.

    PubMed Central  CAS  PubMed  Google Scholar 

  86. Barrett JW, Werden SJ, Wang F, McKillop WM, Jimenez J, Villeneuve D, McFadden G, Dekaban GA: Myxoma virus M130R is a novel virulence factor required for lethal myxomatosis in rabbits. Virus Res. 2009, 144: 258-265. 10.1016/j.virusres.2009.05.009.

    CAS  PubMed  Google Scholar 

  87. Rubartelli A, Poggi A, Sitia R, Zocchi MR: HIV-I Tat: a polypeptide for all seasons. Immunol Today. 1998, 19: 543-545. 10.1016/S0167-5699(98)01351-6.

    CAS  PubMed  Google Scholar 

  88. Zocchi MR, Rubartelli A, Morgavi P, Poggi A: HIV-1 Tat inhibits human natural killer cell function by blocking L-type calcium channels. J Immunol. 1998, 161: 2938-2943.

    CAS  PubMed  Google Scholar 

  89. Opgenorth A, Graham K, Nation N, Strayer D, McFadden G: Deletion analysis of two tandemly arranged virulence genes in myxoma virus, M11L and myxoma growth factor. J Virol. 1992, 66: 4720-4731.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Barrett JW, Sypula J, Wang F, Alston LR, Shao Z, Gao X, Irvine TS, McFadden G: M135R is a novel cell surface virulence factor of myxoma virus. J Virol. 2007, 81: 106-114. 10.1128/JVI.01633-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  91. Colamonici OR, Domanski P, Sweitzer SM, Larner A, Buller RM: Vaccinia virus B18R gene encodes a type I interferon-binding protein that blocks interferon alpha transmembrane signaling. J Biol Chem. 1995, 270: 15974-15978. 10.1074/jbc.270.27.15974.

    CAS  PubMed  Google Scholar 

  92. Symons JA, Alcami A, Smith GL: Vaccinia virus encodes a soluble type I interferon receptor of novel structure and broad species specificity. Cell. 1995, 81: 551-560. 10.1016/0092-8674(95)90076-4.

    CAS  PubMed  Google Scholar 

  93. Marlier D: Vaccination strategies against myxomavirus infections: are we really doing the best?. Tijdschr Diergeneeskd. 2010, 135: 194-198.

    CAS  PubMed  Google Scholar 

  94. Adams MM, van Leeuwen BH, Kerr PJ: Limitations of plasmid vaccines to complex viruses: selected myxoma virus antigens as DNA vaccines were not protective. Vaccine. 2004, 23: 198-204. 10.1016/j.vaccine.2004.05.023.

    CAS  PubMed  Google Scholar 

  95. Willer DO, McFadden G, Evans DH: The complete genome sequence of shope (rabbit) fibroma virus. Virology. 1999, 264: 319-343. 10.1006/viro.1999.0002.

    CAS  PubMed  Google Scholar 

  96. Fenner F, Woodroofe GM: Protection of laboratory rabbits against myxomatosis by vaccination with fibroma virus. Aust J Exp Biol Med Sci. 1954, 32: 653-668. 10.1038/icb.1954.68.

    CAS  PubMed  Google Scholar 

  97. Jerabek J: Applicability of shope fibroma virus replicated in cell cultures for immunoprophylaxis of rabbit myxomatosis. Acta Vet Brno. 1980, 49: 259-267. 10.2754/avb198049030259.

    Google Scholar 

  98. Alfonso M, Pagés-Manté A: Serological response to myxomatosis vaccination by different inoculation systems on farm rabbits. World Rabbit Sci. 2003, 11: 145-156.

    Google Scholar 

  99. Marlier D, Mainil J, Boucraut-Baralon C, Linden A, Vindevogel H: The efficacy of two vaccination schemes against experimental infection with a virulent amyxomatous or a virulent nodular myxoma virus strain. J Comp Pathol. 2000, 122: 115-122. 10.1053/jcpa.1999.0346.

    CAS  PubMed  Google Scholar 

  100. Lavazza A, Graziani M, Tranquillo VM, Botti G, Palotta C, Cerioli M, Capucci L: Serological evaluation of the immunity induced in commercial rabbits by vaccination for myxomatosis and RHD. 2004, Proc. 8th World Rabbit Congress, Puebla, 569-575.

    Google Scholar 

  101. Saurat P, Gilbert Y, Ganiere JP: Study on Myxomatous Virus Modified Strain. Rev Med Vet. 1978, 129: 415-451 (in French).

    Google Scholar 

  102. Petit F, Boucraut-Baralon C, Py R, Bertagnoli S: Analysis of myxoma virus genome using pulsed-field gel electrophoresis. Vet Microbiol. 1996, 50: 27-32. 10.1016/0378-1135(96)00014-4.

    CAS  PubMed  Google Scholar 

  103. Lemière S, Alaphilippe A, Boucher S, Bertagnoli S: Field study of safety and antibody production further to a combined myxomatosis and viral haemorrhagic disease (VHD) vaccination in dwarf rabbits by intradermal route. World Rabbit Sci. 2003, 11: 41-47.

    Google Scholar 

  104. Adams MM, van Leeuwen BH, McFadden G, Kerr PJ: Construction and testing of a novel host-range defective myxoma virus vaccine with the M063 gene inactivated that is non-permissive for replication in rabbit cells. Vet Res. 2008, 39: 60-10.1051/vetres:2008037.

    PubMed  Google Scholar 

  105. Adams MM, van Leeuwen BH, Kerr PJ: Construction and evaluation of live attenuated myxoma virus vaccines with targeted virulence gene deletions. Vaccine. 2008, 26: 5843-5854. 10.1016/j.vaccine.2008.08.036.

    CAS  PubMed  Google Scholar 

  106. Parra F, Prieto M: Purification and characterization of a calicivirus as the causative agent of a lethal hemorrhagic disease in rabbits. J Virol. 1990, 64: 4013-4015.

    PubMed Central  CAS  PubMed  Google Scholar 

  107. Bertagnoli S, Gelfi J, Le Gall G, Boilletot E, Vautherot JF, Rasschaert D, Laurent S, Petit F, Boucraut-Baralon C, Milon A: Protection against myxomatosis and rabbit viral hemorrhagic disease with recombinant myxoma viruses expressing rabbit hemorrhagic disease virus capsid protein. J Virol. 1996, 70: 5061-5066.

    PubMed Central  CAS  PubMed  Google Scholar 

  108. Ferrer M, Negro J: The near extinction of two large European predators: super specialists pay a price. Conserv Biol. 2004, 18: 344-349. 10.1111/j.1523-1739.2004.00096.x.

    Google Scholar 

  109. Gortazar C, Ferroglio E, Hofle U, Frolich K, Vicente J: Diseases shared between wildlife and livestock: a European perspective. Eur J Wildlife Res. 2007, 53: 241-256. 10.1007/s10344-007-0098-y.

    Google Scholar 

  110. Garcia-Bocanegra I, Astorga RJ, Napp S, Casal J, Huerta B, Borge C, Arenas A: Myxomatosis in wild rabbit: design of control programs in Mediterranean ecosystems. Prev Vet Med. 2010, 93: 42-50. 10.1016/j.prevetmed.2009.09.013.

    PubMed  Google Scholar 

  111. Ferreira C, Ramirez E, Castro F, Ferreras P, Alves PC, Redpath S, Villafuerte R: Field experimental vaccination campaigns against myxomatosis and their effectiveness in the wild. Vaccine. 2009, 27: 6998-7002. 10.1016/j.vaccine.2009.09.075.

    PubMed  Google Scholar 

  112. Barcena J, Pages-Mante A, March R, Morales M, Ramirez MA, Sanchez-Vizcaino JM, Torres JM: Isolation of an attenuated myxoma virus field strain that can confer protection against myxomatosis on contacts of vaccinates. Arch Virol. 2000, 145: 759-771. 10.1007/s007050050669.

    CAS  PubMed  Google Scholar 

  113. Angulo E, Bárcena J: Towards a unique and transmissible vaccine against myxomatosis and rabbit haemorrhagic disease for rabbit populations. Wildl Res. 2007, 34: 567-577. 10.1071/WR06160.

    CAS  Google Scholar 

  114. Torres JM, Ramirez MA, Morales M, Barcena J, Vazquez B, Espuna E, Pages-Mante A, Sanchez-Vizcaino JM: Safety evaluation of a recombinant myxoma-RHDV virus inducing horizontal transmissible protection against myxomatosis and rabbit haemorrhagic disease. Vaccine. 2000, 19: 174-182. 10.1016/S0264-410X(00)00183-3.

    CAS  PubMed  Google Scholar 

  115. Morales M, Ramirez MA, Cano MJ, Parraga M, Castilla J, Perez-Ordoyo LI, Torres JM, Barcena J: Genome comparison of a nonpathogenic myxoma virus field strain with its ancestor, the virulent Lausanne strain. J Virol. 2009, 83: 2397-2403. 10.1128/JVI.02189-08.

    PubMed Central  CAS  PubMed  Google Scholar 

  116. Torres JM, Sanchez C, Ramirez MA, Morales M, Barcena J, Ferrer J, Espuna E, Pages-Mante A, Sanchez-Vizcaino JM: First field trial of a transmissible recombinant vaccine against myxomatosis and rabbit hemorrhagic disease. Vaccine. 2001, 19: 4536-4543. 10.1016/S0264-410X(01)00184-0.

    CAS  PubMed  Google Scholar 

  117. Guitton JS, Devillard S, Guenezan M, Fouchet D, Pontier D, Marchandeau S: Vaccination of free-living juvenile wild rabbits (Oryctolagus cuniculus) against myxomatosis improved their survival. Prev Vet Med. 2008, 84: 1-10. 10.1016/j.prevetmed.2007.10.001.

    PubMed  Google Scholar 

  118. Merchant JC, Kerr PJ, Simms NG, Hood GM, Pech RP, Robinson AJ: Monitoring the spread of myxoma virus in rabbit Oryctolagus cuniculus populations on the southern tablelands of New South Wales, Australia. III. Release, persistence and rate of spread of an identifiable strain of myxoma virus. Epidemiol Infect. 2003, 130: 135-147. 10.1017/S0950268802007847.

    PubMed Central  CAS  PubMed  Google Scholar 

  119. Munuswamy-Ramanujam G, Dai E, Liu L, Shnabel M, Sun YM, Bartee M, Lomas DA, Lucas AR: Neuroserpin, a thrombolytic serine protease inhibitor (serpin), blocks transplant vasculopathy with associated modification of T-helper cell subsets. Thromb Haemost. 2010, 103: 545-555. 10.1160/TH09-07-0441.

    CAS  PubMed  Google Scholar 

  120. Chen J, Myllarniemi M, Akyurek LM, Hayry P, Marsden PA, Paul LC: Identification of differentially expressed genes in rat aortic allograft vasculopathy. Am J Pathol. 1996, 149: 597-611.

    PubMed Central  CAS  PubMed  Google Scholar 

  121. Richardson J, Viswanathan K, Lucas A: Serpins, the vasculature, and viral therapeutics. Front Biosci. 2006, 11: 1042-1056. 10.2741/1862.

    CAS  PubMed  Google Scholar 

  122. Bot I, van Berkel TJ, Biessen EA: Viral serine protease inhibitors as anti-atherosclerotic therapy. Curr Opin Investig Drugs. 2007, 8: 729-735.

    CAS  PubMed  Google Scholar 

  123. Dai E, Guan H, Liu L, Little S, McFadden G, Vaziri S, Cao H, Ivanova IA, Bocksch L, Lucas A: Serp-1, a viral anti-inflammatory serpin, regulates cellular serine proteinase and serpin responses to vascular injury. J Biol Chem. 2003, 278: 18563-18572. 10.1074/jbc.M209683200.

    CAS  PubMed  Google Scholar 

  124. Lucas A, Liu L, Dai E, Bot I, Viswanathan K, Munuswamy-Ramunujam G, Davids JA, Bartee MY, Richardson J, Christov A, Wang H, Macaulay C, Poznansky M, Zhong R, Miller L, Biessen E, Richardson M, Sullivan C, Moyer R, Hatton M, Lomas DA, McFadden G: The serpin saga; development of a new class of virus derived anti-inflammatory protein immunotherapeutics. Adv Exp Med Biol. 2009, 666: 132-156. 10.1007/978-1-4419-1601-3_11.

    CAS  PubMed  Google Scholar 

  125. Tardif JC, L'Allier PL, Gregoire J, Ibrahim R, McFadden G, Kostuk W, Knudtson M, Labinaz M, Waksman R, Pepine CJ, Macaulay C, Guertin MC, Lucas A: A randomized controlled, phase 2 trial of the viral serpin Serp-1 in patients with acute coronary syndromes undergoing percutaneous coronary intervention. Circ Cardiovasc Interv. 2010, 3: 543-548. 10.1161/CIRCINTERVENTIONS.110.953885.

    CAS  PubMed  Google Scholar 

  126. Bedard EL, Kim P, Jiang J, Parry N, Liu L, Wang H, Garcia B, Li X, McFadden G, Lucas A, Zhong R: Chemokine-binding viral protein M-T7 prevents chronic rejection in rat renal allografts. Transplantation. 2003, 76: 249-252. 10.1097/01.TP.0000061604.57432.E3.

    CAS  PubMed  Google Scholar 

  127. Dai E, Liu LY, Wang H, McIvor D, Sun YM, Macaulay C, King E, Munuswamy-Ramanujam G, Bartee MY, Williams J, Davids J, Charo I, McFadden G, Esko JD, Lucas AR: Inhibition of chemokine-glycosaminoglycan interactions in donor tissue reduces mouse allograft vasculopathy and transplant rejection. PLoS One. 2010, 5: e10510-10.1371/journal.pone.0010510.

    PubMed Central  PubMed  Google Scholar 

  128. Liu L, Dai E, Miller L, Seet B, Lalani A, Macauley C, Li X, Virgin HW, Bunce C, Turner P, Moyer R, McFadden G, Lucas A: Viral chemokine-binding proteins inhibit inflammatory responses and aortic allograft transplant vasculopathy in rat models. Transplantation. 2004, 77: 1652-1660. 10.1097/01.TP.0000131173.52424.84.

    CAS  PubMed  Google Scholar 

  129. Munuswamy-Ramanujam G, Khan KA, Lucas AR: Viral anti-inflammatory reagents: the potential for treatment of arthritic and vasculitic disorders. Endocr Metab Immune Disord Drug Targets. 2006, 6: 331-343.

    CAS  PubMed  Google Scholar 

  130. Bartee E, McFadden G: Human cancer cells have specifically lost the ability to induce the synergistic state caused by tumor necrosis factor plus interferon-beta. Cytokine. 2009, 47: 199-205. 10.1016/j.cyto.2009.06.006.

    PubMed Central  CAS  PubMed  Google Scholar 

  131. Liu J, Wennier S, Reinhard M, Roy E, Macneill A, McFadden G: Myxoma virus expressing IL-15 fails to cause lethal myxomatosis in European rabbits. J Virol. 2009, 83: 5933-5938. 10.1128/JVI.00204-09.

    PubMed Central  CAS  PubMed  Google Scholar 

  132. Lun X, Yang W, Alain T, Shi ZQ, Muzik H, Barrett JW, McFadden G, Bell J, Hamilton MG, Senger DL, Forsyth PA: Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas. Cancer Res. 2005, 65: 9982-9990. 10.1158/0008-5472.CAN-05-1201.

    PubMed Central  CAS  PubMed  Google Scholar 

  133. Stanford MM, Barrett JW, Nazarian SH, Werden S, McFadden G: Oncolytic virotherapy synergism with signaling inhibitors: Rapamycin increases myxoma virus tropism for human tumor cells. J Virol. 2007, 81: 1251-1260. 10.1128/JVI.01408-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  134. Stanford MM, McFadden G: Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther. 2007, 7: 1415-1425. 10.1517/14712598.7.9.1415.

    CAS  PubMed  Google Scholar 

  135. Wang F, Ma Y, Barrett JW, Gao X, Loh J, Barton E, Virgin HW, McFadden G: Disruption of Erk-dependent type I interferon induction breaks the myxoma virus species barrier. Nat Immunol. 2004, 5: 1266-1274. 10.1038/ni1132.

    CAS  PubMed  Google Scholar 

  136. Woo Y, Kelly KJ, Stanford MM, Galanis C, Shin Chun Y, Fong Y, McFadden G: Myxoma virus is oncolytic for human pancreatic adenocarcinoma cells. Ann Surg Oncol. 2008, 15: 2329-2335. 10.1245/s10434-008-9924-z.

    PubMed  Google Scholar 

  137. Kim M, Williamson CT, Prudhomme J, Bebb DG, Riabowol K, Lee PW, Lees-Miller SP, Mori Y, Rahman MM, McFadden G, Johnston RN: The viral tropism of two distinct oncolytic viruses, reovirus and myxoma virus, is modulated by cellular tumor suppressor gene status. Oncogene. 2010, 29: 3990-3996. 10.1038/onc.2010.137.

    PubMed Central  CAS  PubMed  Google Scholar 

  138. Kim M, Madlambayan GJ, Rahman MM, Smallwood SE, Meacham AM, Hosaka K, Scott EW, Cogle CR, McFadden G: Myxoma virus targets primary human leukemic stem and progenitor cells while sparing normal hematopoietic stem and progenitor cells. Leukemia. 2009, 23: 2313-2317. 10.1038/leu.2009.219.

    PubMed Central  CAS  PubMed  Google Scholar 

  139. Lun XQ, Zhou H, Alain T, Sun B, Wang L, Barrett JW, Stanford MM, McFadden G, Bell J, Senger DL, Forsyth PA: Targeting human medulloblastoma: oncolytic virotherapy with myxoma virus is enhanced by rapamycin. Cancer Res. 2007, 67: 8818-8827. 10.1158/0008-5472.CAN-07-1214.

    PubMed Central  CAS  PubMed  Google Scholar 

  140. Stanford MM, Shaban M, Barrett JW, Werden SJ, Gilbert PA, Bondy-Denomy J, Mackenzie L, Graham KC, Chambers AF, McFadden G: Myxoma virus oncolysis of primary and metastatic B16F10 mouse tumors in vivo. Mol Ther. 2008, 16: 52-59. 10.1038/sj.mt.6300348.

    PubMed Central  CAS  PubMed  Google Scholar 

  141. Rahman MM, Madlambayan GJ, Cogle CR, McFadden G: Oncolytic viral purging of leukemic hematopoietic stem and progenitor cells with Myxoma virus. Cytokine Growth Factor Rev. 2010, 21: 169-175. 10.1016/j.cytogfr.2010.02.010.

    PubMed Central  CAS  PubMed  Google Scholar 

  142. Rosas C, Van de Walle GR, Metzger SM, Hoelzer K, Dubovi EJ, Kim SG, Parrish CR, Osterrieder N: Evaluation of a vectored equine herpesvirus type 1 (EHV-1) vaccine expressing H3 haemagglutinin in the protection of dogs against canine influenza. Vaccine. 2008, 26: 2335-2343. 10.1016/j.vaccine.2008.02.064.

    PubMed Central  CAS  PubMed  Google Scholar 

  143. Rosas CT, Tischer BK, Perkins GA, Wagner B, Goodman LB, Osterrieder N: Live-attenuated recombinant equine herpesvirus type 1 (EHV-1) induces a neutralizing antibody response against West Nile virus (WNV). Virus Res. 2007, 125: 69-78. 10.1016/j.virusres.2006.12.009.

    CAS  PubMed  Google Scholar 

  144. Rosas CT, Konig P, Beer M, Dubovi EJ, Tischer BK, Osterrieder N: Evaluation of the vaccine potential of an equine herpesvirus type 1 vector expressing bovine viral diarrhea virus structural proteins. J Gen Virol. 2007, 88: 748-757. 10.1099/vir.0.82528-0.

    CAS  PubMed  Google Scholar 

  145. Van de Walle GR, May MA, Peters ST, Metzger SM, Rosas CT, Osterrieder N: A vectored equine herpesvirus type 1 (EHV-1) vaccine elicits protective immune responses against EHV-1 and H3N8 equine influenza virus. Vaccine. 2010, 28: 1048-1055. 10.1016/j.vaccine.2009.10.123.

    CAS  PubMed  Google Scholar 

  146. Ruitenberg KM, Walker C, Wellington JE, Love DN, Whalley JM: DNA-mediated immunization with glycoprotein D of equine herpesvirus 1 (EHV-1) in a murine model of EHV-1 respiratory infection. Vaccine. 1999, 17: 237-244. 10.1016/S0264-410X(98)00192-3.

    CAS  PubMed  Google Scholar 

  147. Wellington JE, Lawrence GL, Love DN, Whalley JM: Expression and characterization of equine herpesvirus 1 glycoprotein D in mammalian cell lines. Arch Virol. 1996, 141: 1785-1793. 10.1007/BF01718301.

    CAS  PubMed  Google Scholar 

  148. Reuter JD, Vivas-Gonzalez BE, Gomez D, Wilson JH, Brandsma JL, Greenstone HL, Rose JK, Roberts A: Intranasal vaccination with a recombinant vesicular stomatitis virus expressing cottontail rabbit papillomavirus L1 protein provides complete protection against papillomavirus-induced disease. J Virol. 2002, 76: 8900-8909. 10.1128/JVI.76.17.8900-8909.2002.

    PubMed Central  CAS  PubMed  Google Scholar 

  149. Roberts A, Reuter JD, Wilson JH, Baldwin S, Rose JK: Complete protection from papillomavirus challenge after a single vaccination with a vesicular stomatitis virus vector expressing high levels of L1 protein. J Virol. 2004, 78: 3196-3199. 10.1128/JVI.78.6.3196-3199.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  150. Brandsma JL, Shylankevich M, Su Y, Roberts A, Rose JK, Zelterman D, Buonocore L: Vesicular stomatitis virus-based therapeutic vaccination targeted to the E1, E2, E6, and E7 proteins of cottontail rabbit papillomavirus. J Virol. 2007, 81: 5749-5758. 10.1128/JVI.02835-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  151. Barrett JW, Alston LR, Wang F, Stanford MM, Gilbert PA, Gao X, Jimenez J, Villeneuve D, Forsyth P, McFadden G: Identification of host range mutants of myxoma virus with altered oncolytic potential in human glioma cells. J Neurovirol. 2007, 13: 549-560. 10.1080/13550280701591526.

    CAS  PubMed  Google Scholar 

  152. OIE, Disease distribution maps of myxomatosis. [http://www.oie.int/wahis/public.php?page=disease_status_map&disease_type=Terrestrial&disease_id=90&disease_category_terrestrial=-1&empty=999999&disease_category_aquatic=-1&disease_serotype=0&sta_method=semesterly&selected_start_year=2009&selected_report_period=2&selected_start_month=1&page=disease_status_map&date_submit=OK]

  153. Graham KA, Lalani AS, Macen JL, Ness TL, Barry M, Liu LY, Lucas A, Clark-Lewis I, Moyer RW, McFadden G: The T1/35kDa family of poxvirus-secreted proteins bind chemokines and modulate leukocyte influx into virus-infected tissues. Virology. 1997, 229: 12-24. 10.1006/viro.1996.8423.

    CAS  PubMed  Google Scholar 

  154. Lalani AS, McFadden G: Secreted poxvirus chemokine binding proteins. J Leukoc Biol. 1997, 62: 570-576.

    CAS  PubMed  Google Scholar 

  155. Dai E, Guan H, Liu L, Little S, McFadden G, Vaziri S, Cao H, Ivanova IA, Bocksch L, Lucas A: Serp-1, a viral anti-inflammatory serpin, regulates cellular serine proteinase and serpin responses to vascular injury. J Biol Chem. 2003, 278: 18563-18572. 10.1074/jbc.M209683200.

    CAS  PubMed  Google Scholar 

  156. Lomas DA, Evans DL, Upton C, McFadden G, Carrell RW: Inhibition of plasmin, urokinase, tissue plasminogen activator, and C1S by a myxoma virus serine proteinase inhibitor. J Biol Chem. 1993, 268: 516-521.

    CAS  PubMed  Google Scholar 

  157. Upton C, Macen JL, Wishart DS, McFadden G: Myxoma virus and malignant rabbit fibroma virus encode a serpin-like protein important for virus virulence. Virology. 1990, 179: 618-631. 10.1016/0042-6822(90)90129-F.

    CAS  PubMed  Google Scholar 

  158. Messud-Petit F, Gelfi J, Delverdier M, Amardeilh MF, Py R, Sutter G, Bertagnoli S: Serp2, an inhibitor of the interleukin-1beta-converting enzyme, is critical in the pathobiology of myxoma virus. J Virol. 1998, 72: 7830-7839.

    PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

GM's lab is supported by grants from NIAID (R01 AI080607-01A1), NCI (R01 CA138541-01 and R21 CA149869) and the Bankhead-Coley Foundation (1BT02). GVdW is a postdoctoral fellow of the FWO Flanders. We would also like to thank Jia Liu and Masmudur Rahman for proof-reading the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bart Spiesschaert.

Additional information

Authors' contributions

BS performed an extensive literature study on the subject, analyzed the acquired information and wrote the manuscript; GM and KH both revised the manuscript critically for important intellectual content concerning their own area of expertise; HN was involved in the initial drafting of the manuscript; GVdW participated in its design and coordination, critically revised the manuscript and helped to draft the manuscript. All authors read and approved the final manuscript.

Competing interests

GM is a co-founder of Viron Therapeutics, which is developing viral proteins as therapeutics. The other authors declare that they have no competing interests.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Spiesschaert, B., McFadden, G., Hermans, K. et al. The current status and future directions of myxoma virus, a master in immune evasion. Vet Res 42, 76 (2011). https://doi.org/10.1186/1297-9716-42-76

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1297-9716-42-76

Keywords