Comparative Medicine Copyright 2015 by the American Association for Laboratory Animal Science

Vol 65, No 6 December 2015 Pages 499–507

Overview

Rabbit Models for Studying Human Infectious Diseases Xuwen Peng,* John A Knouse, and Krista M Hernon Using an appropriate animal model is crucial for mimicking human disease conditions, and various facets including genetics, anatomy, and pathophysiology should be considered before selecting a model. Rabbits (Oryctolagus cuniculus) are well known for their wide use in production of antibodies, eye research, atherosclerosis and other cardiovascular diseases. However, a systematic description of the rabbit as primary experimental models for the study of various human infectious diseases is unavailable. This review focuses on the human infectious diseases for which rabbits are considered a classic or highly appropriate model, including AIDS (caused by HIV1), adult T-cell leukemia–lymphoma (human T-lymphotropic virus type 1), papilloma or carcinoma (human papillomavirus) , herpetic stromal keratitis (herpes simplex virus type 1), tuberculosis (Mycobacterium tuberculosis), and syphilis (Treponema pallidum). In addition, particular aspects of the husbandry and care of rabbits used in studies of human infectious diseases are described. Abbreviations: CRPV, cottontail rabbit papillomavirus; HPV, human papillomavirus; HSV1, herpes simplex virus type 1; HTLV1, human T-lymphotropic virus type 1

Rodents, especially mice, are the animals most commonly used as models in biomedical research because of their many advantages, including short lifespan, high reproductive rate, cost-effectiveness, ease of genetic manipulation, and the availability of numerous inbred strains with well-defined genetic backgrounds and of a wide variety of reagents for experiments. However, mice and rats are not susceptible to infection by several human pathogens, including HIV1.132 In addition, the causative mutations resulting in human disease sometimes do not cause corresponding pathologic changes in mice,4 and mice are not always suitable for studies due to their small size and phylogenetic features.104 Alternatively, the domestic rabbit (Oryctolagus cuniculus), especially New Zealand white rabbits, has attracted more attention in recent years in biomedical and pharmaceutical research, especially in cardiovascular diseases, because of its intermediate size and phylogenetic proximity to primates.32,44,104 Furthermore, rabbits have served as the primary experimental model for some human infectious diseases because of their susceptibility to infection and the similarity of pathogenesis to that in humans. With the rapid development of rabbit genomics, proteomics, transgenic and knockout lines, and rabbit-specific reagents,12,111,141 the value of rabbits as experimental models in biomedical research likely will increase, especially in their ability to bridge the gap between rodents and the large animal models often required for preclinical and translational research. Because rabbits used in cardiovascular diseases and other traditional studies have been well described Received: 06 Apr 2015. Revision requested: 03 Jun 2015. Accepted: 28 Jun 2015. Department of Comparative Medicine, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania *Corresponding author. Email: [email protected]

elsewhere,5,30,32,104 the current overview focuses on the rabbit models primarily used to study various human infectious diseases.

Rabbit Models for HIV1 Infection and AIDS

The human AIDS complex is a spectrum of conditions that mainly arises due to infection with the retrovirus HIV1, which primarily is transmitted through unprotected sexual contact, contaminated blood transfusions and needles, and from mother to child.115 The virus has a narrow host range for infection, and the restricted species-specific infection in humans is determined by viral entry into target cells, primarily CD4+ T cell helper lymphocytes. This action is mediated by binding of its trimeric envelope spike protein (gp160) to the primary receptor, human CD4,66 followed by binding to specific coreceptors, including human chemokine receptor CCR5.23,130 In addition, the host-specific barriers to HIV1 infection might be related to various antiviral factors that specifically target viruses to restrict their propagation. For example, members of the tripartite-motif–bearing family of proteins have both host- and retrovirus-specific antiviral properties.120,129 All of these characteristics have made the identification and development of a suitable animal model for the study of HIV1 infection extremely difficult. During the last few decades, considerable progress has been made in understanding the HIV1 infection and in the development of animal models of this disease. However, an immunecompetent and infection-susceptible small-animal model is still needed to complement the existing models for studying HIV1 infection and pathogenesis, as well as for evaluating new drugs and developing vaccines. To date, various NHP species serve as the ultimate model of HIV1 infection, but they are either not readily

499

cm15000043.indd 499

12/3/2015 9:34:09 AM

Vol 65, No 6 Comparative Medicine December 2015

available as endangered species or are subject to ethical and high cost concerns. Even among NHP, chimpanzees (Pan troglodytes) and gibbon apes are the only species that are fully susceptible to HIV1 infection.1 However, some macaque species, especially rhesus monkeys, have been used as ‘gold standards’ for testing vaccines because they can be challenged with chimeric SHIV derived from SIV and HIV1.79 SHIV does not produce the same disease in the monkeys as that of HIV1 infection in humans, and it differs from HIV1 in terms of the requirements for effective vaccination.92 Wild-type mice and rats are well known to be nonpermissive for HIV1 infection.131 Even expression of human CD4, the known principal receptor for HIV1, failed to render mouse cells permissive to viral infection.80 In contrast, compared with rodents, rabbits are relatively more susceptible to HIV1 infection, given that both infected normal rabbit cells as well as infected rabbits themselves have been generated.35,43,70,71,133 However, the rabbit infections were much less efficient than those in human lymphocytes, and infected rabbits failed to show consistent clinical signs of AIDS-like disease. Rabbit CD4 may not play the same role as the human molecule in supporting HIV1 infection of cells; instead, rabbits might have additional nonCD4 HIV1 receptors.46 Sequence comparisons of the CD4 molecule cloned from a rabbit thymus cDNA library showed that 6 of the 18 residues implicated in HIV1 binding differ between the human and rabbit proteins, and no correlation between CD4 expression by rabbit cell lines and their ability to support HIV1 infection was seen.46 A subsequent study demonstrated that expression of human CD4 in rabbit PBMC enhanced their infection by HIV1 and resulted in a rapid depletion of the infected cells.78 Furthermore, 2 groups then simultaneously reported the successful generation of transgenic rabbits bearing human CD4.29,117 One of the groups reported that expression of human CD4 in the transgenic rabbits was restricted to lymphocytes that expressed the rabbit homolog, and peripheral blood lymphocytes from the CD4-transgenic rabbits produced greater amounts of HIVq p24 core protein after HIV1 infection in vitro than did similar lymphocytes from nontransgenic rabbits. However, no study describing HIV1 infection of the transgenic rabbits themselves was reported.117 Other in vitro studies similarly demonstrated enhanced susceptibility of lymphocytes derived from the CD4-transgenic rabbits to HIV1 infection.29 Furthermore, in vivo HIV1 infection of the CD4-transgenic rabbits was confirmed through virus isolation, detection of HIV1-specific DNA in peripheral blood lymphocytes, and seroconversion to various HIV1 proteins. However, the infected transgenic rabbits lacked any clinical sign of AIDS-like disease,29 indicating that these animals were not sufficiently susceptible to HIV1 infection for disease development. Various coreceptors are hypothesized to be involved in susceptibility to HIV1 infection, including the β-chemokine receptor CCR5, which is an essential receptor for HIV1 entry into cells.23,41 Cells from a rabbit cell line (SIRC) that expressed the human forms of CD4 and CCR5 were highly permissive for HIV1 infection, and the level of viral replication in the SIRC cells approached that seen in human cells.118 In addition, HIV1 virions produced in SIRC rabbit cells were able to propagate the infection by successfully infecting human PBMC. These findings suggest that transgenic rabbits expressing the human genes for CD4 and CCR5 may accurately reflect HIV1 replication in vivo and serve as a useful small animal model. However, this long-awaited transgenic rabbit model has not yet been reported.

A more recent study showed promise for the development of a rabbit model by demonstrating the considerable natural HIV1 permissivity of rabbit cells.127 In this study, the HIV1 susceptibility of rabbit cells, including primary T cells and macrophages expressing human CD4 and CCR5, was evaluated. The study showed that the rabbit cells, unlike with mouse and rat cells, could support the functions of the regulatory viral proteins Tat and Rev, the processing of the Gag antigen, and the release of HIV1 particles at the levels comparable to those in human cells. It was further demonstrated that viral particles produced by the rabbit T cells were highly infectious. These results suggest that the host-specific factors hampering HIV1 infection could be evaded through receptor complex transgenesis combined with modifications in the HIV1 gag (and possibly vif) genes, potentially rendered rabbits fully permissive to infection by HIV1.127 With increased understanding of the host-specific factors required for HIV1 infection and the ongoing development of new gene-modification techniques, a genetically modified rabbit model that is highly susceptible to HIV1 infection may eventually become possible. This hope is supported by a recent study that described the successful knock-in of the human CD4 and CCR5 homologs into rabbit embryos through the new technique using transcription activator-like effector nuclease (TALEN).125

Rabbit Model for HTLV1 Infection and Adult T-cell Leukemia–Lymphoma

Human T-lymphotropic virus type 1 (HTLV1) was the first human retrovirus isolated in 1979, the virus was obtained from both the fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma.40,109 HTLV1 is a single-stranded diploid RNA virus that predominantly affects CD4+ lymphocytes. An estimated 20 million people worldwide are infected with HTLV1.110 Approximately 5% of infected persons develop adult T-cell leukemia– lymphoma, and another 1% to 3% develops the condition known as HTLV-associated myelopathy–tropical spastic paraparesis or various other immune-mediated disorders. In contrast, more than 90% of HTLV1-infected persons remain lifelong asymptomatic, persistent carriers.42 This virus can be transmitted through breast milk, sexual contact, intravenous drug use, blood transfusion, and organ transplantation.42 Research in the last 3 decades was directed at understanding the biologic and pathogenic properties of HTLV1 and to the development of various experimental vaccination and therapeutic strategies against HTLV1 infection. However, a licensed vaccine remains unavailable, and the mechanism underlying the neoplastic pathogenesis in patients with adult T-cell leukemia–lymphoma is not yet completely understood. A variety of animal models have been used to study the early events of HTLV1 infection, including viral transmission, pathogenesis, host immunologic response to the infection and development of novel therapies (see reference 27 for review). Rabbits, rats, and mice are used most commonly among these models, of which rabbits and rats (but not mice) can be infected with HTLV1. Although rats can be infected experimentally, further studies found them to be unreliable models to test the early spread of the virus due to considerable interstrain variation in the response to viral infection.47,63 Consequently, rabbits have been extensively used since the 1980s as a model of HTLV1 infection due to their ease of handling and the consistency of viral infection and transmission.

500

cm15000043.indd 500

12/3/2015 9:34:09 AM

Rabbit models of human infectious diseases

Rabbits can be easily infected with HTLV1 after intravenous injection of the MT2 cell line, a T-cell leukemia line derived from a patient with adult T-cell leukemia–lymphoma.89 For studying viral transmission, rabbits inoculated with cell lines are euthanized at defined intervals after infection for the collection of tissue samples, including PBMC, spleen, mesenteric lymph nodes, and gut-associated lymphoid tissues, for further analysis. For example, plasma can be used to detect antibody response by Western blotting, and mononuclear leukocytes can be isolated to detect viral load by real-time PCR analysis or cultured to detect virus expression after exposure.27,53 Rabbits respond similarly to humans infected with HTLV1 in terms of antibody response, and the viral parameters measured in this rabbit model are similar to those used to detect the infection in humans.85,142 Although HTLV1-infected rabbits generally do not develop clinical disease, they simulate the persistent asymptomatic infection manifested by most human patients. Research has shown that persistent HTLV1 infection is determined by a balance between host immune responses and viral spread, and immunomodulatory therapies have been used clinically in HTLV1-infected patients. To understand how the immunosuppressive treatments might influence the host–virus relationship, the effects of cyclosporine (an immunomodulatory drug) was studied in rabbits during early infection of HTLV1.52 The resulting data indicated that immunologic control plays a key role for early HTLV1 spread and has important implications for therapeutic intervention during HTLV1-associated diseases. Collectively, the rabbit model for HTLV1 infection has played indispensable roles in the evaluation of immune responses, parameters of infection, routes of transmission, and viral genetic determinants of infection (by using molecular clones of HTLV1).18,27,51,67,139 The main disadvantage of this model is that the infected rabbits do not spontaneously develop the earlier-described immune-mediated diseases that occur in a small percentage of patients.

Rabbit Models for Human Papillomavirus Infection

Papillomaviruses are a group of small DNA viruses that are associated with infection and neoplasia of cutaneous and mucosal epithelial tissues in both humans and some animal species.13,144 More than 100 types of human papillomavirus (HPV) have been defined genetically. These viruses are classified as either ‘low-risk’ viruses involved in the development of benign genital warts or ‘high risk’ viruses (also called carcinogenic viruses), given their roles in the etiologies of various cutaneous and mucosal cancers, notably cervical cancers.91,144 Among the high-risk HPV16 and HPV18 are the most prevalent HPV associated with cervical cancers. Epidemiologic and molecular studies indicate that, although high-risk (carcinogenic) HPV have been found in more than 90% of cervical cancers, the viruses alone are insufficient to cause malignant transformation; various cofactors (inactivation of tumor suppressor genes, activating mutation of cellular oncogenes, or others) apparently are required.143 In addition, various preliminary studies found that activated H-ras, an oncogene found in many human malignant tumors (including some HPV16- or HPV18-associated cervical cancers),75 was required for malignant transformation of HPV16-immortalized human cervical cells26 and that both HPV16 DNA and EJ-ras were required for transformation of primary cells.88

Due to the restricted species-specificity of these viruses, a natural animal model is not available in which to study the infection and pathogenesis of HPV or to evaluate potential vaccines and therapeutic agents. Current preclinical models of natural papillomaviral infections include those involving rabbits, dogs, and cattle. Because of its similarity in genomic structure and tumorigenesis, cottontail rabbit papillomavirus (CRPV) has long been used as an animal model to study the pathogenesis and development of prophylactic and therapeutic strategies for oncogenic HPV infection.7,10,17,69 The major primary advantages of the CRPV model is the reliable and predictable induction of skin papillomas after treatment with purified CRPV DNA, either applied onto scarified rabbit skin or delivered by gene gun,9,61 thus supporting studies on viral genetics and immunology. To study the interaction between a carcinogenic papillomavirus and potential cofactors in vivo, transgenic rabbits were generated that carried the CRPV genome either alone or with the EJ-ras oncogene.107 Analyses showed that the rabbits that expressed CRPV E6/E7 genes alone developed skin papillomas only, but those expressing both CRPV E6/E7 genes and the EJ-ras oncogene were born with extensive squamous cell carcinomas of the skin.107 In a subsequent study, transgenic rabbits with targeted expression of EJ-ras in the skin due to control by the CRPV upstream regulatory region showed growth of keratoacanthomas during the first week of age.105 The keratoacanthomas were morphologically similar to the tumors in humans and spontaneously regressed at about 2 mo of age. After complete regression of the keratoacanthomas, EJ-ras expression was undetectable, and there was no new tumor growth.105 However, CRPV infection of the skin of 2-mo-old transgenic rabbits (after regression of keratoacanthomas) reinitiated the expression of the EJ-ras transgene and accelerated tumorigenesis compared with that in nontransgenic rabbits.106 Together, these results indicate that an activated oncogene such as EJ-ras might act as a cofactor to increase the tumorigenicity or carcinogenicity of the oncogenic papillomavirus infection. This synergistic effect has been confirmed in the human literature: H-ras mutation was identified in 21% of patients with poorly or moderately differentiated cervical tumors.73 However, the mechanisms underlying the synergistic effect between activated H-ras and oncogenic papillomaviruses during carcinogenesis needs to be elucidated further. To assess host immune response to HPV infection and for preclinical evaluation of initial vaccination and immunotherapeutic testing, animal models of HPV infection are required. However, the narrow species specificity of papillomaviruses makes these studies particularly challenging.34 Species-restrictive barriers prevent the infection by and replication of HPV in all immunocompetent laboratory animals. Among the natural papillomavirus infection models, the CRPV model offers several advantages as a preclinical model for studying host immunity to papillomavirus infection.17 One advantage is that papillomas can be generated by direct infection of skin with naked viral DNA.8,69 This feature provides opportunities for genetic modification of the viral genome by site-directed mutagenesis, which can be used to induce epitopes into the various viral genes for testing specific immunity. The CRPV genome tolerates multiple modifications without losing its ability to induce skin papillomas.56 For example, an HPV16 E7 T-cell epitope can be introduced into the E7 gene of the CRPV genome, such that the modified virus retains full tumorigenicity.59 To assess immune responses to the HPV epitopes in an infection

501

cm15000043.indd 501

12/3/2015 9:34:09 AM

Vol 65, No 6 Comparative Medicine December 2015

model for papillomaviruses, a transgenic rabbit model expressing the HLA-A2.1 gene, a well-characterized human MHC class I gene, has been established.58,59 HLA-A2.1 was expressed and colocalized exclusively with rabbit MHC class I on cell surfaces in all transgenic rabbits. The transgenic rabbits vaccinated with a computer-predicted HLA-A2.1-restricted HPV16 E7 multivalent epitope (amino-acid residues 82 through 90) DNA vaccine showed significant to complete protection against infection with modified CRPV DNA containing an embedded HPV16 E7/82–90 epitope within the CRPV E7 gene.6,59 In addition, these transgenic rabbit models have been used to screen and characterize other computer-predicted HLA-A2.1-restricted epitopes from CRPV E1 as potential DNA vaccines for both protective and therapeutic purposes against CRPV infection.55,57,60 Data from these studies indicate that the HLA-A2.1 transgenic rabbit model might provide opportunities to directly test HLA-A2.1-restricted epitopes of HPV proteins in the context of a human MHC class I gene.

Rabbit Models for Ocular Herpes Infection

Herpes simplex virus type 1 (HSV1) is a double-stranded DNA virus, which is unique in its ability to establish a lifelong latent infection in human hosts.128 Infection with HSV1 frequently is associated with ocular pathology and remains a leading cause of infectious blindness worldwide, with an estimated 40,000 new cases of severe monocular visual impairment or blindness each year.33 Ocular HSV1 is transmitted by close contact and affects about 500,000 people in the United States.33 After initial infection, HSV1 travels by retrograde axonal transport to local sensory ganglia, where it either replicates or establishes latency. When reactivated, the virus travels from the sensory ganglia to the cornea, leading to herpetic stromal keratitis and then infectious blindness mainly due to stromal opacification resulting from tissue damage, edema, and corneal scarring.108 Because ocular herpes infections remain a major health problem and no vaccines have been developed, the current research is focused on understanding the complex immune mechanisms associated with viral reactivation, creating improved therapies, and developing vaccines against both primary and recurrent infections.33 Numerous animal models have been used to investigate the pathogenesis, treatment, and prevention of HSV1-associated diseases for several decades. Although guinea pigs are commonly used for studying genital herpesvirus infection, mice and rabbits are typically used for studies of HSV1 latency, reactivation, and recurrence.134 The advantages and disadvantages for using mice and rabbits in studies of HSV1 infection have been well addressed in a recent review article.134 Rabbits are recognized as a valuable model for studying ocular HSV1 infection, mainly because of their large eyes, thus allowing for easy access to corneal lesions for imaging and quantification by slit-lamp examination and for increased quantities of ocular and neural tissues for assessment. In addition, rabbits have an abundant tear film volume, which facilitates the collection of tears for efficient detection of the infectious virus and DNA.134 Furthermore, herpetic stromal keratitis is reliably induced in rabbits after ocular inoculation with HSV1, and the disease course in rabbits is similar to the human disease, including viral reactivation.48,93,96 Because of these advantages, rabbits have long been used for in vivo experiments in HSV1 latency,97 studies of spontaneous and induced reactivation in the HSV1 latent rabbits, and research on recurrent HSV1-specific corneal lesions and therapeutic in-

tervention.134 For example, one of the earliest studies using New Zealand white rabbits inoculated with the McKrae strain showed that HSV1 remains latent in the trigeminal ganglion between attacks of HSV1 ocular disease.97 In addition, experiments using rabbit models demonstrated that adrenergic neural elements act as a trigger to induce HSV1 reactivation during latency.72,74,116 Compared with mice, rabbits are a more suitable model for studying recurrent HSV1 ocular lesions49,65 64 because of the ease of access for slit-lamp examination, as mentioned earlier.134 In addition, a recent study reported the use of a novel humanized HLA-A2.1 transgenic rabbit model for the preclinical evaluation of vaccines against ocular herpes.14 In that study, immunization of humanized HLA-transgenic rabbits with a mixture of 3 human glycoprotein D lipopeptides resulted in the production of HSV1-specific CD8+ T cells, reduced HSV1 ocular replication, and reduced number of corneal lesion after ocular challenge by HSV1. HLA-transgenic rabbits likely will become a powerful model to study protective immunity induced by prophylactic vaccination against HSV1 infection.

Rabbit Models for Tuberculosis

Tuberculosis is a widespread infectious disease caused by Mycobacterium tuberculosis in humans.76 This bacterium is an aerobic, acid-fast, and gram-positive organism that is spread through the air, usually by coughing and sneezing from people with active infection, and that typically attacks the lungs. Owing to its epidemic status in many parts of the world, tuberculosis remains a major cause of morbidity and mortality, especially in some developing countries. The disease has not been controlled efficiently due to the absence of affordable long-term treatments, emergence of drug resistance, and lack of an effective vaccine.77 The World Health Organization estimates that one-third of the world’s population has been infected with M. tuberculosis, and new infections occur in about 1% of the population each year.138 More specifically, an estimated 9 million new cases occurred throughout the world in 2013, resulting in 1.5 million deaths, most of which were in developing countries.138 Moreover, tuberculosis is reported to have caused about 11% of deaths among adults with AIDS.20 In AIDS patients, mortality may be related to multiorgan effects in sites other than primary to lung infection.100 In immunocompetent people, cell-mediated immunity (T lymphocytes and macrophages), through the formation of granulomas, is regarded as the dominant mode of protection against M. tuberculosis.102 In contrast, granulomas are absent or poorly formed in people with weakened immune responses, especially those infected with HIV.76 Bacteria confined inside granulomas can become dormant and thus associated with latent infection. In addition, a cell-mediated response to M. tuberculosis can create cavities filled with caseous necrotic material, due to abnormal cell death.99,100 The most commonly used animal models in the study of tuberculosis are well addressed in a recent review.24 Among the 3 species reviewed (mice, guinea pigs, and rabbits), mice are relatively resistant to tuberculosis infection, followed by rabbits, whereas guinea pigs were more susceptible to infection than were mice and rabbits. Unlike mice and guinea pigs, rabbits are the only model that develops pulmonary cavitation like that in humans; therefore, this model offers a means for studying the factors leading to this form of disease and to bronchial spread of the pathogen.24 Researches have found that delayed-type hypersensitivity and cell-mediated immunity each play a role in rabbits’ ability

502

cm15000043.indd 502

12/3/2015 9:34:10 AM

Rabbit models of human infectious diseases

to form cavities after exposure to whole tubercle bacilli or mycobacterial protein and lipid components.22,140 Therefore, the rabbit model provides a greater potential for studying the transmission of tuberculosis than do other animal models.24 Despite the advantage of developing lung cavitation seen in humans, rabbits currently available for use in research are relatively resistant to tuberculosis infection. Unfortunately, an inbred strain that was reported to be susceptible to airborne M. tuberculosis84 no longer exists. However, the relative resistance of rabbits to infection allows them to serve as a spontaneous model for studying latent tuberculosis infection in humans.24,123 In contrast, rabbits are significantly susceptible to bovine mycobacterial infection (M. bovis) due to inhaled bovine tubercle bacilli or bronchoscopic inoculation. The resulting pulmonary pathology more closely resembles human M. tuberculosis infection than that seen in mice and guinea pigs.95 Furthermore, infection of rabbits with different strains of M. tuberculosis might result in varied spectrums of disease.86,122 For example, rabbits infected with M. tuberculosis HN878 (a hypervirulent strain of the W-Beijing lineage) developed progressive cavitary disease similar to that seen in humans with active tuberculosis.87 In contrast, infection with the strain CDC 1551 (a hyperimmunogenic clinical isolate) was confined in rabbit lungs as a latent infection; however, unlike in human latent infection, rabbits infected with CDC 1551 tend not to reactivate disease unless they are experimentally immunosuppressed.87 Further study revealed that the rabbits with latent tuberculosis infection showed early activation of T cells and macrophages, as well as an early peak in the level of TNFα,123 and immunosuppression induced by corticosteroids resulted in reactivation of the disease in the rabbits.87 This corticosteroid model is useful, in that it is the only animal model available for studying the immune reconstitution during tuberculosis infection after immunosuppression. For these reasons, New Zealand white rabbits provide a valuable animal model for studying tuberculosis. Effective use of this model might facilitate the elucidation of mechanisms underlying host protective immunity, development of new vaccines, and evaluation of novel diagnostic and treatments.

Rabbit Models for Syphilis

A sexually transmitted infectious disease caused by Treponema pallidum (T. pallidum) subspecies pallidum, syphilis remains to be a significant global health problem, with an estimated 12 million new cases of infection each year.137 T. pallidum is a member of the Spirochaetaceae family of spiral-shaped, gram-negative, highly mobile bacteria.31 This bacterium is characterized by a small genome that lacks the ability to encode sufficient metabolic pathways necessary to make most of its macronutrients.37,99 Therefore, this organism cannot be grown in vitro because it must rely heavily on its host for nutrients, and it cannot survive outside of mammalian cells. Humans are the only natural reservoir for the subspecies pallidum. 73,119 Syphilis is transmitted primarily through sexual contact; however, congenital syphilis can occur due to transmission from mother to fetus during pregnancy or at birth.136 Infection is initiated by the penetration of T. pallidum through dermal microabrations or intact mucous membranes, resulting in primary syphilis. Untreated syphilis progresses through stages including secondary, latent, and tertiary disease. Late syphilis can affect the cardiovascular system and central nervous system of infected persons.62,83,103 In addition, pregnant mothers with untreated syphilis can spread the disease through transplacental

transmission of the bacteria to the unborn infants, resulting in the congenital syphilis.112 Nearly half of the children infected with syphilis during fetal development die shortly before or after birth,98 and adults infected with syphilis have an enhanced risk for HIV transmission and acquisition.15 A recent review paper systematically addresses many questions that remain to be studied regarding syphilis, including the biologic basis of the disease and the development of new tools for diagnosis, treatment and prevention.54 Although humans are the only natural hosts of T. pallidum, rabbits are the only mammal to develop naturally occurring syphilis caused by T. paraluis-cuniculi, a bacterium closely related to T. pallidum with antigenic crossreactivity and similar symptoms.2,21,25,45 These characteristics suggest that rabbits might also be susceptible to experimental infection with T. pallidum. The testes of rabbits were shown to be particularly susceptible to T. pallidum infection.135 Because T. pallidum cannot be grown in culture, this unique property led to the use of rabbits as an in-vivo medium for the propagation of T. pallidum and a primary model for studying the pathogenesis and immunity of human syphilis.65,126 Additional studies have found that infection of rabbits by intradermal inoculation with T. pallidum resulted in development of primary lesions, which resemble the chancre of syphilis histologically and morphologically in humans.114 Experimental syphilis in rabbits usually ended after the resolution of the primary lesions, without development of secondary syphilis lesions, especially when the rabbits were infected with the Nichols strain of T. pallidum. However, a different strain of T. pallidum, Melbourne 1, induces secondary syphilis lesions in rabbits.121 In addition, rabbit models for experimental congenital and neurologic syphilis have been created by intravenous administration of the organism. 38,39,126 However, neither the neuroinvasive nor congenital transmission model has been well characterized.103,126 Because the rabbit model closely reflects human infection, it is continually used to study disease pathogenesis,103 explore new therapies,3,81,82 and develop potential vaccines.11,90

The Husbandry and Care of Rabbits Used to Study Human Infectious Disease

As with all animal models used in research, the husbandry and care of rabbits used to study human infectious diseases requires careful consideration regarding the health status and agent used, unique phenotypes of the animal, handling and restraint, and personnel risk. Rabbits are the most numerous of the research animals covered by the Animal Welfare Act, and their use in research in the United States is regulated by the US Department of Agriculture.131 Owing to its well-established influences on research, the husbandry and care of rabbits is aimed at protecting them from unintended disease exposure and environmental stress.19,101,124 Maintaining an established health status typically minimizes variability in research. In addition to the specific requirements outlined by the Animal Welfare Act for rabbit housing environments and care, the human infectious agents used in a rabbit study dictate additional needs depending on the biosafety level of the agent and the associated animal biosafety level for the in vivo application. This information is concisely outlined in several references, including Biosafety in Microbiologic and Biomedical Laboratories,16 and the agents we have discussed here predominately require Animal Biosafety Level 2 practices.

503

cm15000043.indd 503

12/3/2015 9:34:10 AM

Vol 65, No 6 Comparative Medicine December 2015

With the ongoing development and use of transgenic rabbits in research, attention must be paid to unique phenotypes that are associated with genetic manipulation. These considerations must address not only the overall assessment and housing of the animals but also the methods for handling, restraint, and sampling. Using a calm and confident approach during handling and restraint is most successful and likely will minimize the associated stress and potential for injury. Rabbits have a fragile bone structure that comprises a lightweight skeleton with well-developed, large muscles attached, thus making them prone to fractures or subluxation of the vertebrae.124 To avoid these outcomes, a handler must support both the front and rear ends of a rabbit and prevent it from kicking and struggling. The specific model of human disease and associated study goals also will influence matters of restraint, sampling, and other conditions. For example, rabbit models of HIV1 infection may require the development of specialized restraint devices for blood collection that might be inappropriate or unnecessary for rabbits modeling HPV infection. In addition to the basic risks inherent when working with animal models (for example, allergies and zoonoses, bites and scratches, and ergonomic concerns), additional risks must be assessed when working with human pathogens. All persons working with rabbits should be enrolled in their institution’s occupational health and safety program, to identify and prevent risks to personnel. In addition, they should be appropriately trained in all procedures involved with the animal models and pathogens they might contact. All training and participation in health and safety programs should be documented and updated regularly. More detailed information on this topic can be obtained by using Occupational Health and Safety in the Care and Use of Research Animals.94 Preplanning, training, and using appropriate references and resources are all vital to the successful husbandry and care of rabbits modeling human infectious disease.

Conclusion

In this overview, we have addressed the uses of laboratory rabbits as primary models for studies of several human infectious diseases caused by HIV1, HTLV1, HPV, HSV1, M. tuberculosis, and T. pallidum. As we described earlier, rabbit models have played important roles in elucidating the pathogenesis of these diseases and in the exploration and assessment of therapeutic and protective agents. However, challenges regarding the use of rabbit models remain, especially for immunologic and genetic studies, due to the paucity of immunologic reagents and the as-yet incomplete genomic sequence. Fortunately, considerable progress has been achieved in recent years in light of the genomics and proteomics for this species.12,111 Most interesting, a recent study reported the successful generation of targeted mutations in rabbit embryos and production of knockout rabbits by using a new technique, RNA-guided Cas9 nucleases.141 In addition, the current HLA-A2.1 transgenic rabbits, which have been used successfully in studies of HPV6,58 and HSV1,14 might also be helpful in the testing of vaccines that induce CD8+ cytotoxic T lymphocytes against other human pathogens that are permissive or semipermissive in rabbits, including HTLV1,36,51 adenovirus,113 EBV-like viruses,50 and tuberculosis.28 Rabbit likely will play increasingly important roles in the modeling of human diseases, especially in translational studies, especially upon the completion of the rabbit genomic sequence, increased availability of cytokines and antibodies, and creation of additional lines of genetically modified rabbits.

Acknowledgment

This work was supported by the C Max and Sylvia S Lang Professorship Fund, George T Harrell Professorship Fund, and General Fund in the Department of Comparative Medicine of the Pennsylvania State University College of Medicine. We thank Dr Ronald Wilson for his support of the related work.

References

1. Agy MB, Katze MG. 1993. HIV1 infection of animals: the search for a pathogenesis model continues. AIDS 7 Suppl 1:S37–S42. 2. Baker-Zander SA, Lukehart SA. 1984. Antigenic cross-reactivity between Treponema pallidum and other pathogenic members of the family Spirochaetaceae. Infect Immun 46:116–121. 3. Baker-Zander SA, Lukehart SA. 1989. Efficacy of cefmetazole in the treatment of active syphilis in the rabbit model. Antimicrob Agents Chemother 33:1465–1469. 4. Bosze Z, Hiripi L, Carnwath JW, Hiemann H. 2003. The transgenic rabbit as model for human diseases and as a source of biologically active recombinant proteins. Transgenic Res 12:541–553. 5. Bosze Z, Houdebine LM. 2006. Application of rabbits in biomedical research: a review. World Rabbit Sci 14:1–14. 6. Bounds CE, Hu J, Cladel NM, Balogh K, Christensen ND. 2011. Vaccine generated immunity targets an HPV16 E7 HLA-A2.1-restricted CD8+ T-cell epitope relocated to an early gene or a late gene of the cottontail rabbit papillomavirus (CRPV) genome in HLA–A2.1 transgenic rabbits. Vaccine 29:1194–1200. 7. Brandsma JL. 2005. The cottontail rabbit papillomavirus model of high-risk HPV-induced disease. Methods Mol Med 119:217–235. 8. Brandsma JL, Xiao W. 1993. Infectious virus replication in papillomas induced by molecularly cloned cottontail rabbit papillomavirus DNA. J Virol 67:567–571. 9. Brandsma JL, Yang ZH, Barthold SW, Johnson EA. 1991. Use of a rapid, efficient inoculation method to induce papillomas by cottontail rabbit papillomavirus DNA shows that the E7 gene is required. Proc Natl Acad Sci USA 88:4816–4820. 10. Breitburd F, Salmon J, Orth G. 1997. The rabbit viral skin papillomas and carcinomas: a model for the immunogenetics of HPV-associated carcinogenesis. Clin Dermatol 15:237–247. 11. Brinkman MB, McGill MA, Pettersson J, Rogers A, Matejkova P, Smajs D, Weinstock GM, Norris SJ, Palzkill T. 2008. A novel Treponema pallidum antigen, TP0136, is an outer membrane protein that binds human fibronectin. Infect Immun 76:1848–1857. 12. Broad Institute of MIT and Harvard.[Internet] 2009. Rabbit Genome Project.[Cited 31March 2015]. Available at: http://www.broadinstitute.org/scientific-community/science/projects/mammalsmodels/rabbit/rabbit-genome-project 13. Campo MS. 2002. Animal models of papillomavirus pathogenesis. Virus Res 89:249–261. 14. Chentoufi AA, Dasgupta G, Christensen ND, Hu J, Choudhury ZS, Azeem A, Jester JV, Nesburn AB, Wechsler SL, BenMohamed L. 2010. A novel HLA (HLA-A*0201) transgenic rabbit model for preclinical evaluation of human CD8+ T-cell epitope-based vaccines against ocular herpes. J Immunol 184:2561–2571. 15. Chesson HW, Heffelfinger JD, Voigt RF, Collins D. 2005. Estimates of primary and secondary syphilis rates in pesons with HIV in the United States, 2002. Sex Transm Dis 32:265–269. 16. Chosewood LC, Wilson DE, editors. 2009. Biosafety in microbiological and biomedical laboratories. 5th ed. Washington (DC): US Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention, National Institutes of Health. 17. Christensen ND. 2005. Cottontail rabbit papillomavirus (CRPV) model system to test antiviral and immunotherapeutic strategies. Antivir Chem Chemother 16:355–362. 18. Collins ND, Newbound GC, Ratner L, Lairmore MD. 1996. In vitro CD4+ lymphocyte transformation and infection in a rabbit model with a molecular clone of human T-cell lymphotropic virus type 1. J Virol 70:7241–7246.

504

cm15000043.indd 504

12/3/2015 9:34:10 AM

Rabbit models of human infectious diseases

19. Conour LA, Murray KA, Brown MJ. 2006. Preparation of animals for research—issues to consider for rodents and rabbits. ILAR J 47:283–293. 20. Corbett EL, Watt CJ, Walker N, Maher D, Williams BC, Raviglione MC, Dye C. 2003. The growing burden of tuberculosis global treands and interactions with the HIV epidemic. Arch Intern Med 163:1009–1021. 21. Cunliffe-Beamer TL, Fox RR. 1981. Venereal spirochetosis of rabbits: description and diagnosis. Lab Anim Sci 31:366–371. 22. Dannenberg AM Jr., editor. 2006. Pathogenesis of human pulmonary tuberculosis—insights from the rabbit model. Washington (DC): ASM Press. 23. Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR. 1996. Identification of a major coreceptor for primary isolates of HIV1. Nature 381:661–666. 24. Dharmadhikari AS, Nardell EA. 2008. What animal models teach humans about tuberculosis. Am J Respir Cell Mol Biol 39:503–508. 25. DiGiacomo RF, Lukehart SA, Talburt CD, Baker-Zander SA, Giddens WE Jr, Condon J, Brown CW. 1985. Chronicity of infection with Treponema paraluis-cuniculi in New Zealand white rabbits. Genitourin Med 61:156–164. 26. DiPaolo JA, Woodworth CD, Popescu NC, Notario PV, Doniger J. 1989. Induction of human cervical squamous cell carcinoma by sequential transfection with human papillomavirus 16 DNA and viral Harvey ras. Oncogene 4:395–399. 27. Dodon MD, Villaudy J, Gazzolo L, Haines R, Lairmore M. 2012. What we are learning on HTLV1 pathogenesis from animal models. Front Microbiol 3:320. 28. Dorman SE, Hatem CL, Tyagi S, Aird K, Lopez-Molina J, Pitt ML, Zook BC, Dannenberg AM Jr, Bishai WR, Manabe YC. 2004. Susceptibility to tuberculosis: clues from studies with inbred and outbred New Zealand white rabbits. Infect Immun 72:1700–1705. 29. Dunn CS, Mehtali M, Houdebine LM, Gut J-P, Kirn A, Aubertin A-M. 1995. Human immunodeficiency virus type 1 infection of human CD4-transgenic rabbits. J Gen Virol 76:1327–1336. 30. Duranthon V, Beaujean N, Brunner M, Odening KE, Santos AN, Kacskovics I, Hiripi L, Weinstein EJ, Bosze Z. 2012. On the emerging role of rabbit as human disease model and the instrumental role of novel transgenic tools. Transgenic Res 21:699–713. 31. Eccleston K, Collins L, Higgins SP. 2008. Primary syphilis. Int J STD AIDS 19:145–151. 32. Fan J, Watanabe T. 2003. Transgenic rabbits as therapeutic protein bioreactors and human disease models. Pharmacol Ther 99:261–282. 33. Farooq AV, Shukla D. 2012. Herpes simplex epithelial and stromal keratitis: an epidemiologic update. Surv Ophthalmol 57:448–462. 34. Fausch SC, Da Silva DM, Eiben GL, Le Poole IC, Kast WM. 2003. HPV protein/peptide vaccines: from animal models to clinical trials. Front Biosci 8:s81–s91. 35. Filice G, Cereda PM, Varnier OE. 1988. Infection of rabbits with human immunodeficiency virus. Nature 335:366–369. 36. Franchini G, Tartaglia J, Markham P, Benson J, Fullen J, Wills M, Arp J, Dekaban G, Paoletti E, Gallo RC. 1995. Highly attenuated HTLV type I-env poxvirus vaccines induce protection against a cell-associated HTLV type I challenge in rabbits. AIDS Res Hum Retroviruses 11:307–313. 37. Fraser CM, Norris SJ, Weinstock GM, White O, Sutton GG, Dodson R, Gwinn M, Hickey EK, Clayton R, Ketchum KA, Sodergren E, Hardham JM, McLeod MP, Salzberg S, Peterson J, Khalak H, Richardson D, Howell JK, Chidambaram M, Utterback T, McDonald L, Artiach P, Bowman C, Cotton MD, Fujii C, Garland S, Hatch B, Horst K, Roberts K, Sandusky M, Weidman J, Smith HO, Venter JC. 1998. Complete genome sequence of Treponema pallidum, the syphilis spirochete. Science 281:375–388. 38. Fitzgerald TJ. 1985. Experimental congenital syphilis in rabbits. Can J Microbiol 31:757–762. 39. Froberg MK, Fitzgerald TJ, Hamilton TR, Zarabi M. 1993. Pathology of congenital syphilis rabbits. Infect Immun 61:4743–4749.

40. Gallo RC. 2005. The discovery of the first human retrovirus, HTLV1 and HLTV2. Retrovirology 2:17. 41. Ghorpade A, Xia MQ, Hyman BT, Persidsky Y, Nukuna A, Bock P, Che M, Limoges J, Gendelman HE, Mackay CR. 1998. Role of the β-chemokine receptors CCR3 and CCR5 in human immunodeficiency virus type 1 infection of monocytes and microglia. J Virol 72:3351–3361. 42. Goncalves DU, Proietti FA, Ribas JGR, Araujo MG, Pinheiro SR, Guedes AC, Carneiro-Proietti BF. 2010. Epidemiology, treatment, and prevention of human T-cell leukemia virus type 1-associated diseases. Clin Microbiol Rev 23:577–589. 43. Gordon MR, Truckenmiller ME, Recker DP, Dickerson DR, Kuta E, Kulaga H, Kindt TJ. 1990. Evidence for HIV1 infection in rabbits. A possible model for AIDS. Proc NY Acad Sci 616:270–280. 44. Graur D, Duret L, Gouy M. 1996. Phytogenetic position of the order Lagomorpha (rabbits, hares, and allies). Nature 379:333–335. 45. Graves S. 1980. Susceptibility of rabbits venereally infected with Treponema paraluis-cuniculi to superinfections with Treponema pallidum. Br J Vener Dis 56:387–389. 46. Hague BF, Sawasdikosol S, Brown TJ, Lee K, Recker DP, Kindt TJ. 1992. CD4 and its role in infection of rabbit cell lines by human immunodeficiency virus type 1. Proc Natl Acad Sci USA 89:7963–7967. 47. Hakata Y, Yamada M, Shida H. 2001. Rat CRM1 is responsible for the poor activity of human T-cell leukemia virus type 1 Rex protein in rat cells. J Virol 75:11515–11525. 48. Haruta Y, Maguire LJ, Rootman DS, Hill JM. 1987. Recurrent herpes simplex virus type 1 corneal epithelial lesions after radial keratotomy in the rabbit. Arch Ophthalmol 105:692–694. 49. Haruta Y, Rootman DS, Xie L, Kiritoshi A, Hill JM. 1989. Recurrent HSV1 corneal lesions in rabbits induced by cyclophosphamide and dexamethasone. Invest Ophthalmol Vis Sci 30:371–376. 50. Hayashi K, Jin Z, Onoda S, Joko H, Teramoto N, Oda W, Tanaka T, Liu YX, Koirala TR, Oka T, Kondo E, Yoshino T, Takahashi K, Akagi T. 2003. Rabbit model for human EBV-associated hemophagocytic syndrome (HPS): sequential autopsy analysis and characterization of IL2-dependent cell lines established from herpesvirus papio-induced fatal rabbit lymphoproliferative diseases with HPS. Am J Pathol 162:1721–1736. 51. Haynes RA 2nd, Phipps AJ, Yamamoto B, Green P, Laimore MD. 2009. Development of a cytotoxic T-cell assay in rabbits to evaluate early immune response to human T-lymphotropic virus type 1 infection. Viral Immunol 22:397–405. 52. Haynes RA 2nd, Ware E, Premanandan C, Zimmerman B, Yu L, Phipps AJ, Laimore MD. 2010. Cyclosporine-induced immune suppression alters establishment of HTLV1 infection in a rabbit model. Blood 115:815–823. 53. Haynes RA 2nd, Zimmerman B, Millward L, Ware E, Premanandan C, Yu L, Phipps AJ, Lairmore MD. 2010. Early spatial and temporal events of human T-lymphotropic virus type 1 spread following bloodborne transmission in a rabbit model of infection. J Virol 84:5124–5130. 54. Ho EL, Lukehart SA. 2011. Syphilis: using modern approaches to understand an old disease. J Clin Invest 121:4584–4592. 55. Hu J, Budgeon LR, Balogh KK, Peng X, Cladel NM, Christensen ND. 2014. Long-peptide therapeutic vaccination against CRPVinduced papillomas in HLA-A2.1 transgenic rabbits. Trials Vaccinol 3:134–142. 56. Hu J, Cladel NM, Balogh K, Christensen ND. 2007. Impact of genetic changes to the CRPV genome and their application to the study of pathogenesis in vivo. Virology 358:384–390. 57. Hu J, Cladel N, Peng X, Balogh K, Christensen ND. 2008. Protective immunity with an E1 multivalent epitope DNA vaccine against cottontail rabbit papillomavirus (CRPV) infection in an HLA-A2.1 transgenic rabbit model. Vaccine 26:809–816. 58. Hu J, Peng X, Budgeon LR, Cladel NM, Balogh KK, Christensen ND. 2007. Establishment of a cottontail rabbit papillomaviru/HLAA2.1-transgenic rabbit model. J Virol 81:7171–7177. 59. Hu J, Peng X, Schell TD, Budgeon LR, Cladel NM, Christensen ND. 2006. An HLA-A2.1-transgenic rabbit model to study immunity to papillomavirus infection. J Immunol 177:8037–8045.

505

cm15000043.indd 505

12/3/2015 9:34:10 AM

Vol 65, No 6 Comparative Medicine December 2015

60. Hu J, Schell TD, Peng X, Cladel NM, Balogh KK, Christensen ND. 2010. Using HLA-A2.1 transgenic rabbit model to screen and characterize new HLA-A2.1 restricted epitope DNA vaccines. J Vaccines Vaccin 1:1. 61. Ito Y, Evans CA. 1961. Induction of tumors in domestic rabbits with nucleic acid preparations from partially purified Shope papilloma virus and from extracts of the papillomas of domestic and cottontail rabbits. J Exp Med 114:485–500. 62. Jackman JD Jr, Radolf JD. 1989. Cardiovascular syphilis. Am J Med 87:425–433. 63. Kannagi M, Ohashi T, Hanabuchi S, Kato H, Koya Y, Hasegawa A, Masuda T, Yoshki T. 2000. Immunological aspects of rat models of HTLV- type 1-infected T lymphoproliferative disease. AIDS Res Hum Retroviruses 16:1737–1740. 64. Kaufman HE, Varnell ED, Gebhardt BM, Thompson HW, Hill JM. 1996. Propanolol suppression of ocular HSV1 recurrence and associated corneal lesions following spontaneous reactivation in the rabbit. Curr Eye Res 15:680–684. 65. Kemp JE, Fitzgerald EM. 1938. Studies in experimental congenital syphilis and the transfer of immunity from immune syphilitic female rabbits to their offspring. J Invest Dermatol 1:353–365. 66. Klatzmann D, Champagne E, Chamaret S, Gruest J, Guetard D, Hercend T, Gluckman JC, Montagnier L. 1984. T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature 312:767–768. 67. Kotani S, Yoshimoto S, Yamato K, Fujishita M, Yamashita M, Ohtsuki Y, Taguchi H, Miyoshi I. 1986. Serial transmission of human T-cell leukemia virus type I by blood transfusion in rabbits and its prevention by use of X-irridiated stored blood. Int J Cancer 37:843–847. 68. Kreider JW, Bartlett GL. 1985. Shope rabbit papilloma–carcinoma complex a model system of HPV infection. Clin Dermatol 3:20–26. 69. Kreider JW, Cladel NM, Patrick SD, Welsh PA, DiAngelo SL, Bower JM, Christensen ND. 1995. High-efficiency induction of papillomas in vivo using recombinant cottontail rabbit papillomavirus DNA. J Virol Methods 55:233–244. 70. Kulaga H, Folks TM, Rutledge R, Kindt TJ. 1988. Infection of rabbit T-cell and macrophage lines with human immunodeficiency virus. Proc Natl Acad Sci USA 85:4455–4459. 71. Kulaga H, Folks TM, Rutledge R, Truckenmiller ME, Gugel E, Kindt TJ. 1989. Infection of rabbits with human immunodeficiency virus 1. A small-animal model for acquired immunodeficiency syndrome. J Exp Med 169:321–326. 72. Kwon BS, Gangarosa LP, Burch KD, deBack J, Hill JM. 1981. Induction of ocular herpes simplex virus shedding by iontophoresis of epinephrine into rabbit cornea. Invest Ophthalmol Vis Sci 21:442–449. 73. Lafond RE, Lukehart SA. 2006. Biological basis for syphilis. Clin Microbiol Rev 19:29–49. 74. Laibson PR, Kibrick S. 1966. Reactivation of herpetic keratitis by epinephrine in rabbit. Arch Ophthalmol 75:254–260. 75. Landro ME, Dalbert D, Picconi MA, Cuneo N, Gonzalez J, Vornetti S, Bazan G, Mural J, Basiletti J, Teyssie AR, Alonio LV. 2008. Human papillomavirus and mutated H-ras oncogene in cervical carcinomas and pathological negative pelvic lymph nodes: a retrospective follow-up. J Med Virol 80:694–701. 76. Lawn SD, Butera ST, Shinnick TM. 2002. Tuberculosis unleashed: the impact of human immunodeficiency virus infection on the host granulomatous response to Mycrobacterium tuberculosis. Microbes Infect 4:635–646. 77. Lawn SD, Zumla AI. 2011. Tuberculosis. Lancet 378:57–72. 78. Leno M, Hague BF, Teller R, Kindt TJ. 1995. HIV1 mediates rapid apoptosis of lymphocytes from human CD4 transgenic but not normal rabbits. Virology 213:450–454. 79. Li J, Lord CI, Haseltine W, Letvin NL, Sodroski J. 1992. Infection of cynomolgus monkeys with a chimeric HIV1–SIVmac virus that expresses the HIV1 envelope glycoproteins. J Acquir Immune Defic Syndr 5:639–646. 80. Lores P, Boucher V, Mackay C, Pla M, Von Boehmer H, Jami J, Barre-Sinoussi F, Weill J-C. 1992. Expression of human CD4 in

81. 82. 83.

84.

85. 86.

87.

88.

89.

90.

91.

92. 93. 94. 95.

96.

97. 98.

transgenic mice does not confer sensitivity to human immunodeficiency virus infection. AIDS Res Hum Retroviruses 8:2063–2071. Lukehart SA, Baker-Zander SA. 1987. Roxithromycin (RU 965): effective therapy for experimental syphilis infection in rabbits. Antimicrob Agents Chemother 31:187–190. Lukehart SA, Fohn MJ, Baker-Zander SA. 1990. Efficacy of azithromycin for therapy of active syphilis in the rabbit model. J Antimicrob Chemother 25 Suppl A:91–99. Lukehart SA, Hook EW 3rd, Baker-Zander SA, Collier AC, Critchlow CW, Handsfield HH. 1988. Invasion of the central nervous system by Treponema pallidum: implications for diagnosis and treatment. Ann Intern Med 109:855–862. Lurie MB, Abramson S, Heppleston AG. 1952. On the response of genetically resistant and susceptible rabbits to the quantitative inhalation of human type tubercle bacilli and the nature of resistance to tuberculosis. J Exp Med 95:119–134. Lydy SL, Conner ME, Marriott SJ. 1998. Relationship between antiTax antibody responses and cocultivatable virus in HTL1-infected rabbits. Virology 250:60–66. Manabe YC, Dannenberg AM Jr, Tyagi SK, Hatem CL, Yoder M, Woolwine SC, Zook BC, Pitt ML, Bishai WR. 2003. Different strains of Mycobacterium tuberculosis cause various spectrums of disease in the rabbit model of tuberculosis. Infect Immun 71:6004–6011. Manabe YC, Kesavan AK, Lopez-Molina J, Hatem CL, Brooks M, Fujiwara R, Hochstein K, Pitt ML, Tufariello J, Chan J, McMurray DN, Bishai WR, Dannenberg AM Jr, Mendez S. 2008. The aerosol rabbit model of TB latency, reactivation and immune reconstitution inflammatory syndrome. Tuberculosis (Edinb) 88:187–196. Matlashewski G, Schneider J, Banks L, Jones N, Murray A, Crawford L. 1987. Human papillomavirus type 16 DNA cooperates with activated ras in transforming primary cells. EMBO J 6:1741–1746. Miyoshi I, Yoshimoto S, Kubonishi I, Fujishita M, Ohtsuki Y, Yamashita M, Yamato K, Hirose S, Taguchi H, Niiya K, Kobayashi M. 1985. Infectious transmission of human T-cell leukemia virus to rabbits. Int J Cancer 35:81–85. Morgan CA, Lukehart SA, Van Voorhis WC. 2002. Immunization with the N-terminal portion of Treponema pallidum repeat protein K attenuates syphilitic lesion development in the rabbit model. Infect Immun 70:6811–6816. Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X, Shah KV, Snijders PJF, Meijer CJLM. 2003. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527. Nath BM, Schumann KE, Boyer JD. 2000. The chimpanzee and other nonhuman-primate models in HIV1 vaccine research. Trends Microbiol 8:426–431. Naito J, Mott KR, Osorio N, Jin L, Perng GC. 2005. Herpes simplex virus type 1 immediate-early protein ICP0 diffuses out of infected rabbit corneas. J Gen Virol 86:2979–2988. National Research Council. 1997. Occupational health and safety in the care and use of research animals. Washington (DC): National Academies Press. Nedeltchev GG, Raghunand TR, Jassal MS, Lun S, Cheng QJ, Bishai WR. 2008. Extrapulmonary dissemination of Mycobacterium bovis but not Mycobacterium tuberculosis in a bronchoscopic rabbit model of cavitary tuberculosis. Infect Immun 77:598–603. Nesburn AB, Bettahi I, Dasgupta G, Chenoufi AA, Zhang X, You S, Morishige N, Wahlert AJ, Brown DJ, Jester JV, Wechsler SL, BenMohamed L. 2007. Functional Foxp3+ CD4+ CD25(Bright+) ‘natural’ regulatory T cells are abundant in rabbit conjunctiva and suppress virus-specific CD4+ and CD8+ effector T cells during ocular herpes infection. J Virol 81:7647–7661. Nesburn AB, Cook ML, Stevens JG. 1972. Latent herpes simplex virus. Isolation from rabbit trigeminal ganglia between episodes of recurrent ocular infection. Arch Ophthalmol 88:412–417. Newman L, Kamb M, Hawkes S, Gomez G, Say L, Seuc A, Broutet N. 2013. Global estimates of syphilis in pregnancy and associated

506

cm15000043.indd 506

12/3/2015 9:34:10 AM

Rabbit models of human infectious diseases

99. 100. 101. 102. 103. 104. 105.

106. 107. 108. 109.

110. 111. 112. 113. 114. 115. 116. 117.

118.

119. 120.

121.

adverse outcomes: analysis of multinational antenatal surveillance data. PLoS Med 10:e1001396. Norris SJ. 1993. Polypeptides of Treponema pallidum: progress toward understanding their structural, functional, and immunologic roles. Treponema pallidum Polypeptide Research Group. Microbiol Rev 57:750–779. North RJ, Jung YJ. 2004. Immunity to tuberculosis. Annu Rev Immunol 22:599–623. Oratz M, Walker C, Schreiber SS, Gross S, Rothschild MA. 1967. Albumin and fibrinogen metabolism in heat- and cold-stressed rabbits. Am J Physiol 213:1341–1349. Paige C, Bishai WR. 2010. Penitentiary or penthouse condo: the tuberculous granuloma from the microbe’s point of view. Cell Microbiol 12:301–309. Peeling RW, Hook EW 3rd. 2005. The pathogenesis of syphilis: the great mimicker, revisited. J Pathol 208:224–232. Peng X. 2012. Transgenic rabbit models for studies of human cardiovascular diseases. Comp Med 62:472–479. Peng X, Griffith JW, Han R, Lang CM, Kreider JW. 1999. Development of keratoacanthomas and squamous cell carcinomas in transgenic rabbits with targeted expression of EJras oncogene in epidermis. Am J Pathol 155:315–324. Peng X, Griffith JW, Lang CM. 2001. Reinitiated expression of EJras transgene in targeted epidermal cells of transgenic rabbits by cottontail rabbit papillomavirus infection. Cancer Lett 171:193–200. Peng X, Olson RO, Christian CB, Lang CM, Kreider JW. 1993. Papillomas and carcinomas in transgenic rabbits carrying EJ-ras DNA and cottontail rabbit papillomavirus DNA. J Virol 67:1698–1701. Pepose JS, Keadle TL, Morrison LA. 2006. Ocular herpes simplex: changing epidemiology, emerging disease patterns, and the potential of vaccine prevention and therapy. Am J Ophthalmol 141:547–557.e2. Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Minna JD, Gallo RC. 1980. Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci USA 77:7415–7419. Proietti FA, Carneiro-Proietti AB, Catalan-Soares BC, Murphy EL. 2005. Global epidermiology of HTLV1 infection and associated diseases. Oncogene 24:6058–6068. Rabbit Genome Biology Network. [Internet] 2014. RGB-Net: a Collaborative European Network on Rabbit Genome Biology. [Cited 31 March 2015]. Available at: http://www.biocomp.unibo.it/rabbit/index.php. Risser WL, Hwang LY. 1996. Problems in the current case definitions of congenital syphilis. J Pediatr 129:499–505. Romanowski EG, Yates KA, Gordon YJ. 2002. Topical corticosteroids of limited potency promote adenovirus replication in the Ad5/NZW rabbit ocular model. Cornea 21:289–291. Sell S, Norris SJ. 1983. The biology, pathology and immunology of syphilis. Int Rev Exp Pathol 24:203–276. Sepkowitz KA. 2001. AIDS-the 20 years. N Engl J Med 344:1764–1772. Shimomura Y, Gangarosa LP Sr, Kataoka M, Hill JM. 1983. HSV1 shedding by iontophoresis of 6 hydroxydopamine followed by tropical epinephrine. Invest Ophthalmol Vis Sci 24:1588–1594. Snyder BW, Vitale J, Milos P, Gosselin J, Gillespie F, Ebert K, Hague BF, Kindt TJ, Wadsworth S, Leibowitz P. 1995. Developmental and tissue-specific expression of human CD4 in transgenic rabbits. Mol Reprod Dev 40:419–428. Speck RF, Penn ML, Wimmer J, Esser U, Hague BF, Kindt TJ, Atchison RT, Goldsmith MA. 1998. Rabbit cells expressing human CD4 and human CCR5 are highly permissive for human immunodeficiency virus type 1 infection. J Virol 72:5728–5734. Stamm LV. 2009. Global challenge of antibiotic-resistant Treponema pallidum. Antimicrob Agents Chemother 54:583–589. Stremlau M, Perron M, Lee M, Li Y, Song B, Javanbakht H, DiazGriffero F, Anderson DJ, Sundquist WI, Sodroski J. 2006. Specific recognition and accelerated uncoading of retroviral capsids by the TRIM5α restriction factor. Proc Natl Acad Sci USA 103:5514–5519. Strugnell RA, Drummond L, Faine S. 1986. Secondary lesions in rabbits experimentally infected with Treponema pallidum. Genitourin Med 62:4–8.

122. Subbian S, Bandyopadhyay N, Tsenova L, O’Brien P, Khetani V, Kushner NL, Peixoto B, Soteropoulos P, Bader JS, Karakousis PC, Fallows D, Kaplan G. 2013. Early innate immunity determines outcome of Mycobacterium tuberculosis pulmonary infection in rabbits. Cell Commun Signal 11:60. 123. Subbian S, Tsenova L, O’Brien P, Yang G, Kushner NL, Parsons S, Peixoto B, Fallows D, Kaplan G. 2012. Spontaneous latency in a rabbit model of pulmonary tuberculosis. Am J Pathol 181:1711–1724. 124. Suckow MA, Stevens KA, Wilson RP. 2012. The laboratory rabbit, guinea pig, hamster, and other rodents. Amsterdam: Elsevier Academic. 125. Tang C, Zhang Q, Li X, Fan N, Yang Y, Quan L, Lai L. 2014. Targeted modification of CCR5 gene in rabbits by TALEN. Yi Chuan 36:360–368. 126. Tantalo LC, Lukehart SA, Marra CM. 2005. Treponema pallidum strain-specific differences in neuroinvasion and clinical phenotype in a rabbit model. J Infect Dis 191:75–80. 127. Tervo H-M, Keppler OT. 2010. High natural permissivity of primary rabbit cells for HIV1, with a viron infectivity defect in macrophages as the final replication barrier. J Virol 84:12300–12314. 128. Toma HS, Murina AT, Areaux RG Jr, Neuman DM, Bhattacharjee PS, Foster TP, Kaufman HE, Hill JM. 2008. Ocular HSV1 latency, reactivation, and recurrent disease. Semin Ophthalmol 23:249–273. 129. Towers GJ. 2007. The control of viral infection by tripartite motif proteins and cyclophilin A. Retrovirology 4:40. 130. Trkola A, Dragic T, Arthos J, Binley JM, Olson WC, Allaway GP, Cheng-Mayer C, Robinson J, Maddon PJ, Moore JP. 1996. CD4dependent, antibody-sensitive interactions between HIV1 and its coreceptor CCR5. Nature 384:184–187. 131. US Department of Agriculture. 2002. US rabbit industry profile. Washington (DC): US Government Printing Office. 132. van Maanen M, Sutton RE. 2003. Rodent models for HIV1 infection and disease. Curr HIV Res 1:121–130. 133. Varnier OE, Kindt TJ. 1992. Current status of HIV1 infection in the rabbit. AIDS Res Hum Retroviruses 8:533–535. 134. Webre JM, Hill JM, Nolan NM, Clement C, McFerrin HE, Bhattacharjee PS, Hsia V, Neumann DM, Foster TP, Lukiw WJ, Thompson HW. 2012. Rabbit and mouse models of HSV1 latency, reactivation, and recurrent eye diseases. J Biomed Biotechnol 2012:612316. 135. Wicher V, Wicher K. 1990. Immunocompetence of inflammatory cells in rabbit testes infected with Treponema pallidum. J Reprod Immunol 18:105–115. 136. Woods CR. 2009. Congenital syphilis-persisting pestilence. Pediatr Infect Dis J 28:536–537. 137. World Health Organization. [Internet] 2011. Prevalence and incidence of selected sexually transmitted infections Chlamydia trachomatis, Neiseria gonorrhoeae, syphilis, and Trichomonas vaginalis: methods and results used by WHO to generate 2005 estimates. [Cited 19 January 2015]. Available at: http://who.int/reproductivehealth/ publications/rtis/9789241502450/en/. 138. World Health Organization. [Internet] 2014. Global tuberculosis report 2014. [Cited 24 December 2014] Available at: http://www. who.int/tb/publications/global_report/en/. 139. Yamade I, Ishiguro T, Seto A. 1991. Infection without antibody response in mother-to-child transmission of HTLV1 in rabbits. J Med Virol 33:268–272. 140. Yamamura Y, Maeda H, Ogawa Y, Hashimoto T. 1986. Experimental pulmonary cavity formation by mycobacterial components and synthetic adjuvants. Microbiol Immunol 30:1175–1187. 141. Yang D, Xu J, Zhu T, Fan J, Lai L, Zhang J, Chen YE. 2014. Effective gene targeting in rabbits using RNA-guided Cas9 nucleases. J Mol Cell Biol 6:97–99. 142. Zhao TM, Hague B, Caudell DL, Simpson RM, Kindt TJ. 2005. Quantification of HTLV1 proviral load in experimentally infected rabbits. Retrovirology 2:34. 143. zur Hausen H. 1991. Viruses in human cancers. Science 254:1167– 1173. 144. zur Hausen H. 2002. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2:342–350.

507

cm15000043.indd 507

12/3/2015 9:34:11 AM

Rabbit Models for Studying Human Infectious Diseases.

Using an appropriate animal model is crucial for mimicking human disease conditions, and various facets including genetics, anatomy, and pathophysiolo...
NAN Sizes 1 Downloads 13 Views