1
|
Mustaffa-Kamal F, Liu H, Pedersen NC, Sparger EE. Characterization of antiviral T cell responses during primary and secondary challenge of laboratory cats with feline infectious peritonitis virus (FIPV). BMC Vet Res 2019; 15:165. [PMID: 31118053 PMCID: PMC6532132 DOI: 10.1186/s12917-019-1909-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
Background Feline infectious peritonitis (FIP) is considered highly fatal in its naturally occurring form, although up to 36% of cats resist disease after experimental infection, suggesting that cats in nature may also resist development of FIP in the face of infection with FIP virus (FIPV). Previous experimental FIPV infection studies suggested a role for cell-mediated immunity in resistance to development of FIP. This experimental FIPV infection study in specific pathogen free (SPF) kittens describes longitudinal antiviral T cell responses and clinical outcomes ranging from rapid progression, slow progression, and resistance to disease. Results Differences in disease outcome provided an opportunity to investigate the role of T cell immunity to FIP determined by T cell subset proliferation after stimulation with different viral antigens. Reduced total white blood cell (WBC), lymphocyte and T cell counts in blood were observed during primary acute infection for all experimental groups including cats that survived without clinical FIP. Antiviral T cell responses during early primary infection were also similar between cats that developed FIP and cats remaining healthy. Recovery of antiviral T cell responses during the later phase of acute infection was observed in a subset of cats that survived longer or resisted disease compared to cats showing rapid disease progression. More robust T cell responses at terminal time points were observed in lymph nodes compared to blood in cats that developed FIP. Cats that survived primary infection were challenged a second time to pathogenic FIPV and tested for antiviral T cell responses over a four week period. Nine of ten rechallenged cats did not develop FIP or T cell depletion and all cats demonstrated antiviral T cell responses at multiple time points after rechallenge. Conclusions In summary, definitive adaptive T cell responses predictive of disease outcome were not detected during the early phase of primary FIPV infection. However emergence of antiviral T cell responses after a second exposure to FIPV, implicated cellular immunity in the control of FIPV infection and disease progression. Virus host interactions during very early stages of FIPV infection warrant further investigation to elucidate host resistance to FIP.
Collapse
Affiliation(s)
- Farina Mustaffa-Kamal
- Department of Medicine and Epidemiology, University of California, One Shields Avenue, Davis, CA, 95616, USA. .,Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Hongwei Liu
- Center for Companion Animal Health, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Niels C Pedersen
- Department of Medicine and Epidemiology, University of California, One Shields Avenue, Davis, CA, 95616, USA.,Center for Companion Animal Health, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Ellen E Sparger
- Department of Medicine and Epidemiology, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
2
|
Pistello M, Conti F, Vannucci L, Freer G. Novel approaches to vaccination against the feline immunodeficiency virus. Vet Immunol Immunopathol 2010; 134:48-53. [PMID: 19896725 DOI: 10.1016/j.vetimm.2009.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Inadequate antigen presentation and/or suboptimal immunogenicity are considered major causes in the failure of human immunodeficiency vaccine to adequately protect against wild-type virus. Several approaches have been attempted to circumvent these hurdles. Here we reviewed some recent vaccinal strategies tested against the feline immunodeficiency virus and focused on: (i) improving antigen presentation by taking advantage of the exquisite ability of dendritic cells to process and present immunogens to the immune system; (ii) boosting immune responses with vaccinal antigens presented in a truly native conformation by the natural target cells of infection. Significance of the studies, possible correlates of protection involved, and implications for developing anti-human immunodeficiency virus vaccines are discussed.
Collapse
Affiliation(s)
- Mauro Pistello
- Retrovirus Center and Virology Section, Department of Experimental Pathology, University of Pisa, Pisa, Italy.
| | | | | | | |
Collapse
|
3
|
Maksaereekul S, Dubie RA, Shen X, Kieu H, Dean GA, Sparger EE. Vaccination with vif-deleted feline immunodeficiency virus provirus, GM-CSF, and TNF-alpha plasmids preserves global CD4 T lymphocyte function after challenge with FIV. Vaccine 2009; 27:3754-65. [PMID: 19464559 PMCID: PMC2802579 DOI: 10.1016/j.vaccine.2009.03.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 03/24/2009] [Accepted: 03/26/2009] [Indexed: 12/17/2022]
Abstract
Feline immunodeficiency virus (FIV) DNA vaccine approaches that included a vif-deleted FIV provirus (FIV-pPPRDeltavif) and feline cytokine expression plasmids were tested for immunogenicity and efficacy by immunization of specific pathogen free cats. Vaccine protocols included FIV-pPPRDeltavif plasmid alone; a combination of FIV-pPPRDeltavif DNA and feline granulocyte macrophage-colony stimulating factor (GM-CSF) and tumor necrosis factor (TNF)-alpha expression plasmids; or a combination of FIV-pPPRDeltavif and feline interleukin (IL)-15 plasmids. Cats immunized with FIV-pPPRDeltavif, GM-CSF and TNF-alpha plasmids demonstrated an increased frequency of FIV-specific T cell proliferation responses compared to other vaccine groups. Immunization with FIV-pPPRDeltavif and IL-15 plasmids was distinguished from other vaccine protocols by the induction of antiviral antibodies. Suppression of virus loads was not observed for any of the FIV-pPPRDeltavif DNA vaccine protocols after challenge with the FIV-PPR isolate. However, prior immunization with FIV-pPPRDeltavif, GM-CSF, and TNF-alpha plasmids resulted in preservation of CD4 T cell functions, including mitogen-induced cytokine expression and antigen-specific proliferation upon infection with FIV. These findings justify further examination of cytokine combinations as adjuvants for lentiviral DNA vaccines.
Collapse
Affiliation(s)
- Saipiroon Maksaereekul
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616
| | - Robert A. Dubie
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616
| | - Xiaoying Shen
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616
| | - Hung Kieu
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616
| | - Gregg A. Dean
- Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27606
| | - Ellen E. Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616
| |
Collapse
|
4
|
Dubie RA, Maksaereekul S, Shacklett BL, Lemongello D, Cole KS, Villinger F, Blozis SA, Luciw PA, Sparger EE. Co-immunization with IL-15 enhances cellular immune responses induced by a vif-deleted simian immunodeficiency virus proviral DNA vaccine and confers partial protection against vaginal challenge with SIVmac251. Virology 2009; 386:109-21. [PMID: 19193388 PMCID: PMC3640844 DOI: 10.1016/j.virol.2009.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 10/30/2008] [Accepted: 01/08/2009] [Indexed: 11/18/2022]
Abstract
Simian immunodeficiency virus (SIV) infection of rhesus macaques is a valuable animal model for human immunodeficiency virus (HIV)-1 vaccine development. Our laboratory recently described the immunogenicity and limited efficacy of a vif-deleted SIVmac239 proviral DNA (SIV/CMVDelta vif) vaccine. The current report characterizes immunogenicity and efficacy for the SIV/CMVDelta vif proviral DNA vaccine when co-inoculated with an optimized rhesus interleukin (rIL)-15 expression plasmid. Macaques co-inoculated with rIL-15 and SIV/CMVDelta vif proviral plasmids showed significantly improved SIV-specific CD8 T cell immunity characterized by increased IFN-gamma ELISPOT and polyfunctional CD8 T cell responses. Furthermore, these animals demonstrated a sustained suppression of plasma virus loads after multiple low dose vaginal challenges with pathogenic SIVmac251. Importantly, SIV-specific cellular responses were greater in immunized animals compared to unvaccinated controls during the initial 12 weeks after challenge. Taken together, these findings support the use of IL-15 as an adjuvant in prophylactic anti-HIV vaccine strategies.
Collapse
Affiliation(s)
- Robert A. Dubie
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Saipiroon Maksaereekul
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Donna Lemongello
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Kelly S. Cole
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Francois Villinger
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322
| | | | - Paul A. Luciw
- Center for Comparative Medicine, University of California, Davis, CA 95616
| | - Ellen E. Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616
| |
Collapse
|
5
|
Reggeti F, Bienzle D. Alloimmunity does not protect from challenge with the feline immunodeficiency virus. Vet Immunol Immunopathol 2008; 124:152-62. [PMID: 18471896 DOI: 10.1016/j.vetimm.2008.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 02/27/2008] [Accepted: 03/07/2008] [Indexed: 10/22/2022]
Abstract
Immune responses against polymorphic host molecules incorporated into lentiviral envelopes during cell budding have induced protection against primate immunodeficiency virus infection. Dendritic cells (DCs) express high levels of MHC molecules and are infectable by lentiviruses. Therefore, in this pilot study we addressed the hypothesis that immunization of cats with allogeneic DC would induce immune responses that protect against challenge with the feline immunodeficiency virus. Two groups of 3 cats each received 3 subcutaneous injections of allogeneic or autologous DC, and were then challenged with viruses propagated in the immunizing DC. Infection status and lymphocyte parameters of cats were assessed during 6 weeks after challenge. MHC II antigens were incorporated into viral particles as identified by Western blot; and antibodies reactive with MHC class II antigens were detected in the serum of cats immunized with allogeneic but not autologous DC. After challenge, all cats had proviral DNA in blood leukocytes from 2 weeks post-challenge onward and seroconverted. Cats immunized with allogeneic DC maintained higher total and CD21(+) lymphocyte concentrations, and higher CD4(+)/CD8(+) lymphocyte ratios; however, these differences were not significantly different from cats that received autologous DC immunizations. Plasma viral load was not significantly different between groups of cats (p=0.204). These results suggest that immunization of cats with allogeneic DC does not induce protective immunity against FIV infection.
Collapse
Affiliation(s)
- F Reggeti
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | | |
Collapse
|
6
|
Sparger EE, Dubie RA, Shacklett BL, Cole KS, Chang WL, Luciw PA. Vaccination of rhesus macaques with a vif-deleted simian immunodeficiency virus proviral DNA vaccine. Virology 2008; 374:261-72. [PMID: 18261756 DOI: 10.1016/j.virol.2008.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 11/12/2007] [Accepted: 01/15/2008] [Indexed: 12/22/2022]
Abstract
Studies in non-human primates, with simian immunodeficiency virus (SIV) and simian/human immunodeficiency virus (SHIV) have demonstrated that live-attenuated viral vaccines are highly effective; however these vaccine viruses maintain a low level of pathogenicity. Lentivirus attenuation associated with deletion of the viral vif gene carries a significantly reduced risk for pathogenicity, while retaining the potential for virus replication of low magnitude in the host. This report describes a vif-deleted simian immunodeficiency virus (SIV)mac239 provirus that was tested as an attenuated proviral DNA vaccine by inoculation of female rhesus macaques. SIV-specific interferon-gamma enzyme-linked immunospot responses of low magnitude were observed after immunization with plasmid containing the vif-deleted SIV provirus. However, vaccinated animals displayed strong sustained virus-specific T cell proliferative responses and increasing antiviral antibody titers. These immune responses suggested either persistent vaccine plasmid expression or low level replication of vif-deleted SIV in the host. Immunized and unvaccinated macaques received a single high dose vaginal challenge with pathogenic SIVmac251. A transient suppression of challenge virus load and a greater median survival time was observed for vaccinated animals. However, virus loads for vaccinated and unvaccinated macaques were comparable by twenty weeks after challenge and overall survival curves for the two groups were not significantly different. Thus, a vif-deleted SIVmac239 proviral DNA vaccine is immunogenic and capable of inducing a transient suppression of pathogenic challenge virus, despite severe attenuation of the vaccine virus.
Collapse
Affiliation(s)
- Ellen E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | | | | | | | | | | |
Collapse
|
7
|
Pistello M. Should accessory proteins be structural components of lentiviral vaccines? Lessons learned from the accessory ORF-A protein of FIV. Vet Immunol Immunopathol 2008; 123:144-9. [PMID: 18304653 DOI: 10.1016/j.vetimm.2008.01.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The FIV regulatory protein Rev and accessory proteins Vif and ORF-A are essential for efficient viral replication and full-blown pathogenesis. Expressed at very low level during viral replication, they are nevertheless processed for recognition by cytotoxic T-lymphocytes (CTLs) and trigger cellular immune responses in FIV-infected cats. The observation that the accessory ORF-A protein of FIV is continuously expressed during viral replication and targeted by cellular immune responses in natural FIV infection, prompted us to investigate the protective potential of this protein. To this aim cats were immunized with three different strategies (protein alone in alum adjuvant, DNA alone, or DNA prime-protein boost) and generated clearly detectable immune responses. Upon challenge with ex vivo homologous FIV, ORF-A immunized cats showed distinct enhancement of acute-phase infection possibly due to an increased expression of the FIV receptor CD134. However, at subsequent sampling points plasma viremia was reduced and CD4+ T-lymphocytes in the circulation declined more slowly in ORF-A immunized than in control animals. These findings support the contention that a multicomponent vaccine, with the inclusion of both accessory and structural proteins, can not only improve the host's ability to control lentivirus replication and slow down disease progression but also draw attention to the fact that even simple immunogens that eventually contribute to protective activity can transiently exacerbate subsequent lentiviral infections.
Collapse
Affiliation(s)
- Mauro Pistello
- Retrovirus Center and Virology Section, Department of Experimental Pathology, University of Pisa, Pisa I-56127, Italy.
| |
Collapse
|
8
|
Shen X, Leutenegger CM, Stefano Cole K, Pedersen NC, Sparger EE. A feline immunodeficiency virus vif-deletion mutant remains attenuated upon infection of newborn kittens. J Gen Virol 2007; 88:2793-2799. [PMID: 17872533 DOI: 10.1099/vir.0.83268-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This report characterizes lentivirus attenuation associated with a vif mutation by inoculation of newborn kittens with a vif-deleted feline immunodeficiency virus provirus plasmid (FIV-pPPRDeltavif). Virus in peripheral blood, antiviral antibody or CD4 T-cell count alterations were not detected in kittens inoculated with FIV-pPPRDeltavif plasmid, with the exception of one kitten that demonstrated FIV Gag antibody production at 42 weeks after inoculation. In contrast, wild-type FIV-pPPR-infected kittens were viraemic, seropositive and exhibited a decrease in the CD4 T-cell subset in peripheral blood. Interestingly, FIV-specific T-cell proliferative responses detected at 32 and 36 weeks after infection were comparable for both FIV-pPPRDeltavif- and wild-type FIV-pPPR-inoculated kittens and suggested the possibility of a discreet tissue reservoir supporting sustained FIV-pPPRDeltavif expression or replication. Overall, these findings confirmed that the severe virus attenuation for both replication and pathogenicity exhibited by a vif-deleted FIV mutant is similar for both neonatal and adult hosts.
Collapse
Affiliation(s)
- Xiaoying Shen
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Christian M Leutenegger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Kelly Stefano Cole
- Department of Medicine, Infectious Diseases Division, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Niels C Pedersen
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Ellen E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
9
|
Lecollinet S, Richardson J. Vaccination against the feline immunodeficiency virus: the road not taken. Comp Immunol Microbiol Infect Dis 2007; 31:167-90. [PMID: 17706778 DOI: 10.1016/j.cimid.2007.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2007] [Indexed: 11/28/2022]
Abstract
Natural infection of domestic cats by the feline immunodeficiency virus (FIV) causes acquired immunodeficiency syndrome (AIDS). FIV is genetically related to human immunodeficiency virus (HIV), and the clinical and biological features of infections caused by feline and human viruses in their respective hosts are highly analogous. Although the obstacles to vaccinating against FIV and HIV would seem to be of comparable difficulty, a licensed vaccine against feline AIDS is already in widespread use in several countries. While this seemingly major advance in prevention of AIDS would appear to be highly instructive for HIV vaccine development, its message has not been heeded by investigators in the HIV field. This review endeavours to relate what has been learned about vaccination against feline AIDS, and to suggest what this may mean for HIV vaccine development.
Collapse
Affiliation(s)
- Sylvie Lecollinet
- UMR 1161 Virologie INRA-AFSSA-ENVA, Ecole Nationale Vétérinaire d'Alfort, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France
| | | |
Collapse
|
10
|
Abstract
Since feline immunodeficiency virus (FIV) was first isolated, international research efforts have been directed towards developing a protective vaccine, not least because it may provide a model for a candidate human immunodeficiency virus (HIV) vaccine. This article reviews the challenges facing vaccine development, the current state of knowledge and future prospects for FIV vaccination.
Collapse
Affiliation(s)
- M J Hosie
- Retrovirus Research Laboratory, Institute for Comparative Medicine, Faculty of Veterinary Medicine, The University of Glasgow
| | | |
Collapse
|
11
|
Abstract
Many experimental strategies have been adopted in experiments to protect cats from FIV infection by vaccination, and some have been successful. The interest in developing a vaccine arose both because FIV is a common cause of morbidity and mortality in pet cats and because the feline virus provides a model for its counterpart in man, human immunodeficiency virus (HIV), for which an effective vaccine is urgently required to halt the current tragic pandemic of acquired immunodeficiency syndrome (AIDS). Shortly after the discovery of FIV and its characterization as a lentivirus, attempts were made to produce a vaccine and success was soon achieved with relatively simple inactivated virus or inactivated virus-infected cell vaccines.82 Further development of this approach led to the introduction in 2002 of the first commercial vaccine against FIV.59 With an estimated prevalence of the infection of up to 25% in populations of pet cats, an effective FIV vaccine could have a significant influence on animal welfare. In addition, this success poses the question of whether a similar strategy might produce an effective vaccine against HIV.
Collapse
|
12
|
Abstract
Animal models for human immunodeficiency virus (HIV) infection play a key role in understanding the pathogenesis of AIDS and the development of therapeutic agents and vaccines. As the only lentivirus that causes an immunodeficiency resembling that of HIV infection, in its natural host, feline immunodeficiency virus (FIV) has been a unique and powerful model for AIDS research. FIV was first described in 1987 by Niels Pedersen and co-workers as the causative agent for a fatal immunodeficiency syndrome observed in cats housed in a cattery in Petaluma, California. Since this landmark observation, multiple studies have shown that natural and experimental infection of cats with biological isolates of FIV produces an AIDS syndrome very similar in pathogenesis to that observed for human AIDS. FIV infection induces an acute viremia associated with Tcell alterations including depressed CD4 :CD8 T-cell ratios and CD4 T-cell depletion, peripheral lymphadenopathy, and neutropenia. In later stages of FIV infection, the host suffers from chronic persistent infections that are typically self-limiting in an immunocompetent host, as well as opportunistic infections, chronic diarrhea and wasting, blood dyscracias, significant CD4 T-cell depletion, neurologic disorders, and B-cell lymphomas. Importantly, chronic FIV infection induces a progressive lymphoid and CD4 T-cell depletion in the infected cat. The primary mode of natural FIV transmission appears to be blood-borne facilitated by fighting and biting. However, experimental infection through transmucosal routes (rectal and vaginal mucosa and perinatal) have been well documented for specific FIV isolates. Accordingly, FIV disease pathogenesis exhibits striking similarities to that described for HIV-1 infection.
Collapse
|
13
|
Yamamoto JK, Pu R, Sato E, Hohdatsu T. Feline immunodeficiency virus pathogenesis and development of a dual-subtype feline-immunodeficiency-virus vaccine. AIDS 2007; 21:547-63. [PMID: 17314517 DOI: 10.1097/qad.0b013e328013d88a] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Gupta S, Leutenegger CM, Dean GA, Steckbeck JD, Cole KS, Sparger EE. Vaccination of cats with attenuated feline immunodeficiency virus proviral DNA vaccine expressing gamma interferon. J Virol 2006; 81:465-73. [PMID: 17079309 PMCID: PMC1797444 DOI: 10.1128/jvi.00815-06] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A feline immunodeficiency virus (FIV) provirus with a vif gene deletion (FIVDelta vifATGgamma) that coexpresses feline gamma interferon (IFN-gamma) was tested as a proviral DNA vaccine to extend previous studies showing efficacy with an FIV-pPPRDelta vif DNA vaccine. Cats were vaccinated with either FIVDelta vifATGgamma or FIV-pPPRDelta vif proviral plasmid DNA or with both FIV-pPPRDelta vif DNA and a feline IFN-gamma expression plasmid (pCDNA-IFNgamma). A higher frequency of FIV-specific T-cell proliferation responses was observed in cats immunized with either FIVDelta vifATGgamma or FIV-pPPRDelta vif plus pCDNA-IFNgamma, while virus-specific cytotoxic-T-lymphocyte responses were comparable between vaccine groups. Antiviral antibodies were not observed postvaccination. Virus-specific cellular and humoral responses were similar between vaccine groups after challenge with a biological FIV isolate (FIV-PPR) at 13 weeks postimmunization. All vaccinated and unvaccinated cats were infected after FIV-PPR challenge and exhibited similar plasma virus loads. Accordingly, inclusion of plasmids containing IFN-gamma did not enhance the efficacy of FIV-pPPRDelta vif DNA immunization. Interestingly, the lack of protection associated with FIV-pPPRDelta vif DNA immunization contrasted with findings from a previous study and suggested that multiple factors, including timing of FIV-pPPRDelta vif inoculations and challenge, as well as route of challenge virus delivery, may significantly impact vaccine efficacy.
Collapse
Affiliation(s)
- Soumi Gupta
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | |
Collapse
|
15
|
Pistello M, Bonci F, Flynn JN, Mazzetti P, Isola P, Zabogli E, Camerini V, Matteucci D, Freer G, Pelosi P, Bendinelli M. AIDS vaccination studies with an ex vivo feline immunodeficiency virus model: analysis of the accessory ORF-A protein and DNA as protective immunogens. J Virol 2006; 80:8856-68. [PMID: 16940498 PMCID: PMC1563914 DOI: 10.1128/jvi.00397-06] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Determining which antigen must be included in AIDS vaccines to confer maximum protection is of utmost importance. In primate models, vaccines consisting of or including accessory viral proteins have yielded conflicting results. We investigated the protective potential of the accessory protein ORF-A of feline immunodeficiency virus (FIV) in cats. All three immunization strategies used (protein alone in alum adjuvant, DNA alone, or DNA prime-protein boost) clearly generated detectable immune responses. Upon challenge with ex vivo homologous FIV, ORF-A-immunized cats showed distinct enhancement of acute-phase infection relative to mock-immunized animals given alum or empty vector DNA. This effect was tentatively attributed to increased expression of the FIV receptor CD134 that was observed in the immunized cats. However, at subsequent sampling points that were continued for up to 10 months postchallenge, the average plasma viral loads of the ORF-A-immunized animals were slightly but consistently reduced relative to those of the control animals. In addition, CD4(+) T lymphocytes in the circulation system declined more slowly in immunized animals than in control animals. These findings support the contention that immunization with lentiviral accessory proteins can improve the host's ability to control virus replication and slow down disease progression but also draw attention to the fact that even simple immunogens that eventually contribute to protective activity can transiently exacerbate subsequent lentiviral infections.
Collapse
Affiliation(s)
- Mauro Pistello
- Dipartimento di Patologia Sperimentale, Università di Pisa, Via San Zeno 37, I-56127 Pisa, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Steckbeck JD, Grieser HJ, Sturgeon T, Taber R, Chow A, Bruno J, Murphy-Corb M, Montelaro RC, Cole KS. Dynamic evolution of antibody populations in a rhesus macaque infected with attenuated simian immunodeficiency virus identified by surface plasmon resonance. J Med Primatol 2006; 35:248-60. [PMID: 16872288 PMCID: PMC3361734 DOI: 10.1111/j.1600-0684.2006.00173.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Increasing evidence suggests that an effective AIDS vaccine will need to elicit broadly neutralizing antibody responses. However, the mechanisms of antibody-mediated neutralization have not been defined. Previous studies from our lab have identified significant differences in the rates of antibody binding to trimeric SIV envelope proteins that correlate with neutralization sensitivity. Importantly, these results demonstrate differences in monoclonal antibody (MAb) binding to neutralization-sensitive and neutralization-resistant envelope proteins, suggesting that one mechanism for virus neutralization may be related to the stability of antibody binding. To date, little has been done to evaluate the binding properties of polyclonal serum antibodies elicited by SIV infection or vaccination. METHODS In the current study, we translate these findings with MAbs to study antibody binding properties of polyclonal serum antibody responses generated in rhesus macaques infected with attenuated SIV. Quantitative and qualitative binding properties of well-characterized longitudinal serum samples to trimeric, recombinant SIV gp140 envelope proteins were analyzed using surface plasmon resonance (SPR) technology (Biacore). RESULTS Results from these studies identified two antibody populations in most of the samples analyzed; one antibody population exhibited fast association/dissociation rates (unstable) while the other population demonstrated slower association/dissociation rates (stable). Over time, the percentage of the total binding response of each antibody population evolved, demonstrating a dynamic evolution of the antibody response that was consistent with the maturation of antibody responses defined using our standard panel of serological assays. However, the current studies provided a higher resolution analysis of polyclonal antibody binding properties, particularly with respect to the early time-points post-infection (PI), that is not possible with standard serological assays. More importantly, the increased stability of the antibody population with time PI corresponded with potent neutralization of homologous SIV in vitro. CONCLUSIONS These results suggest that the stability of the antibody-envelope interaction may be an important mechanism of serum antibody virus neutralization. In addition, measurements of the 'apparent' rates of association and dissociation may offer unique numerical descriptors to characterize the level of antibody maturation achieved by candidate vaccine strategies capable of eliciting broadly neutralizing antibody responses.
Collapse
Affiliation(s)
- J D Steckbeck
- Department of Medicine, Infectious Diseases Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dunham SP, Bruce J, Klein D, Flynn JN, Golder MC, MacDonald S, Jarrett O, Neil JC. Prime-boost vaccination using DNA and whole inactivated virus vaccines provides limited protection against virulent feline immunodeficiency virus. Vaccine 2006; 24:7095-108. [PMID: 17049683 DOI: 10.1016/j.vaccine.2006.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 04/03/2006] [Accepted: 07/03/2006] [Indexed: 10/24/2022]
Abstract
Protection against feline immunodeficiency virus (FIV) has been achieved using a variety of vaccines notably whole inactivated virus (WIV) and DNA. However protection against more virulent isolates, typical of those encountered in natural infections, has been difficult to achieve. In an attempt to improve protection against virulent FIV(GL8), we combined both DNA and WIV vaccines in a "prime-boost" approach. Thirty cats were divided into four groups receiving vaccinations and one unvaccinated control group. Following viral challenge, two vaccinated animals, one receiving DNA alone and one the prime-boost vaccine remained free of viraemia, whilst all controls became viraemic. Animals vaccinated with WIV showed apparent early enhancement of infection at 2 weeks post challenge (pc) with higher plasma viral RNA loads than control animals or cats immunised with DNA alone. Despite this, animals vaccinated with WIV or DNA alone showed significantly lower proviral loads in peripheral blood mononuclear cells and mesenteric lymph node cells, whilst those receiving the DNA-WIV prime-boost vaccine showed significantly lower proviral loads in PBMC, than control animals, at 35 weeks pc. Therefore both DNA and WIV vaccines conferred limited protection against viral challenge but the combination of WIV and DNA in a prime-boost approach appeared to offer no significant advantage over either vaccine alone.
Collapse
Affiliation(s)
- Stephen P Dunham
- Retrovirus Research Laboratory, Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Bearsden Road, Glasgow G61 1QH, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Feline immunodeficiency virus (FIV) is a natural infection of domestic cats, which produces a disease with many similarities to human immunodeficiency virus (HIV) infection in man. The virus is an important cause of morbidity and mortality in pet cats worldwide. As such an effective vaccine is desirable both for its use in veterinary medicine and also as a model for the development of an HIV vaccine. A large number of candidate vaccines have been tested against feline immunodeficiency virus. These include inactivated virus and infected cell vaccines, DNA and viral vectored vaccines, subunit and peptide vaccines and vaccines using bacterial vectors. Ultimately, the development of inactivated virus and infected cell vaccines led to the release of the first licensed vaccine against FIV, in 2002. This review highlights some of the difficulties associated with the development of lentiviral vaccines and some of the lessons that have been learned in the FIV model that are of particular relevance to the development of HIV vaccines.
Collapse
Affiliation(s)
- Stephen P Dunham
- Retrovirus Research Laboratory, Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Bearsden Road, Glasgow G61 1QH, United Kingdom.
| |
Collapse
|
19
|
Gupta S, Leutenegger C, Dean G, Sparger E. Construction and characterization of feline immunodeficiency virus proviral mutants that coexpress interferon gamma and green fluorescent protein. AIDS Res Hum Retroviruses 2006; 22:342-9. [PMID: 16623638 DOI: 10.1089/aid.2006.22.342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vif deletion mutants of the feline immunodeficiency virus (FIV) were designed to express either enhanced green fluorescent protein (EGFP) (FIVdeltavifATGgfp) or feline interferon-gamma (IFN-gamma) (FIVdeltavifATGgamma) by insertion of the nonviral gene into the deletion site of the viral vif gene. Two in-frame start codons within vif were mutated without altering the overlapping pol translation frame to enhance expression of inserted genes. Expression of EGFP and IFN-gamma from FIVdeltavifATGgfp and FIVdeltavifATGgamma proviruses, respectively, was confirmed by fluorescent microscopy and immunocytochemical assays, respectively. Replication of viruses generated from these proviruses was detectable but severely restricted when compared to that of wild-type (WT) FIV-pPPR. A previous study demonstrated induction of protection against homologous FIV challenge by vaccination of cats with an attenuated FIV-pPPRdeltavif proviral DNA vaccine (Lockridge K et al.: Virology 2000;273:67-79). Coexpression of IFN-gamma or other cytokines from this attenuated provirus provides the opportunity to evaluate the ability of an immunomodulator to enhance the safety and efficacy of an infectious attenuated DNA vaccine. Moreover, a vif-deleted FIV provirus that coexpresses a reporter gene such as EGFP may be used to examine the localization of vif mutant viruses in vivo.
Collapse
Affiliation(s)
- Soumi Gupta
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616, USA
| | | | | | | |
Collapse
|
20
|
Busch M, Abel K, Li J, Piatak M, Lifson JD, Miller CJ. Efficacy of a SHIV 89.6 proviral DNA vaccine against mucosal SIVmac239 challenge. Vaccine 2005; 23:4036-47. [PMID: 15963361 DOI: 10.1016/j.vaccine.2005.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2004] [Revised: 02/21/2005] [Accepted: 03/03/2005] [Indexed: 11/23/2022]
Abstract
Sixty percent of rhesus macaques infected with virulence attenuated virus SHIV 89.6 are protected from subsequent intravaginal challenge with pathogenic SIVmac239 [Abel K, Compton L, Rourke T, Montefiori D, Lu D, Rothaeusler K, et al. Simian-human immunodeficiency virus SHIV89.6-induced protection against intravaginal challenge with pathogenic SIVmac239 is independent of the route of immunization and is associated with a combination of cytotoxic T-lymphocyte and alpha interferon responses. J Virol 2003;77(5):3099-118; Miller CJ, McChesney MB, Lu X, Dailey PJ, Chutkowski C, Lu D, et al. Rhesus macaques previously infected with simian/human immunodeficiency virus (HIV) are protected from vaginal challenge with pathogenic SIVmac239. J Virol 1997;71(3):1911-21]. Previously, we have shown that inoculation with a proviral plasmid encoding SHIV 89.6 (pMA SHIV-89.6) results in systemic infection that is delayed compared to SHIV 89.6 virus inoculation [Busch M, Lu D, Fritts L, Lifson JD, Miller CJ. Comparison of virology and immunology in SHIV 89.6 proviral DNA and virus-inoculated rhesus macaques. J Med Primatol 2003;32(4-5):240-6]. We now report that, although monkeys inoculated with pMA SHIV-89.6 or SHIV 89.6 virus had similar plasma anti-SIV binding antibody titers and number of anti-SIV IFN-gamma secreting cells on the day of mucosal SIVmac239 challenge, a smaller proportion of monkeys immunized with pMA SHIV-89.6 were protected from vaginal SIVmac239 challenge compared to monkeys immunized using SHIV 89.6 virus. Protected DNA immunized monkeys had stronger anti-SIV IFN-gamma ELISPOT responses in the acute stage post-challenge than unprotected monkeys. Plasma anti-SIV binding antibody titers and PBMC cytokine responses in the acute stages post-challenge were similar in DNA vaccinated-protected and DNA vaccinated-unprotected monkeys. These results suggest that the delay in systemic infection resulting from delivery of SHIV 89.6 as a plasmid decreased the effectiveness of this live attenuated vaccine.
Collapse
Affiliation(s)
- Marc Busch
- California National Primate Research Center, University California-Davis, County Road 98, Hutchison Drive, Davis, CA 95616, USA
| | | | | | | | | | | |
Collapse
|
21
|
Pistello M, Bonci F, Isola P, Mazzetti P, Merico A, Zaccaro L, Matteucci D, Bendinelli M. Evaluation of feline immunodeficiency virus ORF-A mutants as candidate attenuated vaccine. Virology 2005; 332:676-90. [PMID: 15680433 DOI: 10.1016/j.virol.2004.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 11/19/2004] [Accepted: 12/04/2004] [Indexed: 11/17/2022]
Abstract
Feline immunodeficiency virus (FIV) made defective in the accessory gene ORF-A were previously shown to be greatly attenuated in its ability to replicate in lymphocytes but to grow normally or near normally in other cell types. Here, we examined whether FIV thus mutated could protect specific pathogen-free cats against challenge with ex vivo fully virulent homologous virus. No reversion of the vaccinating infections to wild type ORF-A was noted over 22 months of in vivo infection. Following challenge, 6/6 unvaccinated control cats became readily and heavily infected. In contrast, 3/9 vaccinees showed no evidence of the challenge virus over a 15-month observation period. In the other vaccinees, the challenge virus was predominant for various periods of time, but pre-existing viral loads and CD4 lymphocyte counts were either unaffected or altered only marginally and transiently. These findings show that ORF-A-defective FIV should be further examined as a candidate live attenuated vaccine.
Collapse
Affiliation(s)
- M Pistello
- Department of Experimental Pathology, Retrovirus Center and Virology Section, University of Pisa, Via San Zeno, 37, Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Craigo JK, Li F, Steckbeck JD, Durkin S, Howe L, Cook SJ, Issel C, Montelaro RC. Discerning an effective balance between equine infectious anemia virus attenuation and vaccine efficacy. J Virol 2005; 79:2666-77. [PMID: 15708986 PMCID: PMC548432 DOI: 10.1128/jvi.79.5.2666-2677.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Among the diverse experimental vaccines evaluated in various animal lentivirus models, live attenuated vaccines have proven to be the most effective, thus providing an important model for examining critical immune correlates of protective vaccine immunity. We previously reported that an experimental live attenuated vaccine for equine infectious anemia virus (EIAV), based on mutation of the viral S2 accessory gene, elicited protection from detectable infection by virulent virus challenge (F. Li et al., J. Virol. 77:7244-7253, 2003). To better understand the critical components of EIAV vaccine efficacy, we examine here the relationship between the extent of virus attenuation, the maturation of host immune responses, and vaccine efficacy in a comparative study of three related attenuated EIAV proviral vaccine strains: the previously described EIAV(UK)DeltaS2 derived from a virulent proviral clone, EIAV(UK)DeltaS2/DU containing a second gene mutation in the virulent proviral clone, and EIAV(PR)DeltaS2 derived from a reference avirulent proviral clone. Inoculations of parallel groups of eight horses resulted in relatively low levels of viral replication (average of 10(2) to 10(3) RNA copies/ml) and a similar maturation of EIAV envelope-specific antibody responses as determined in quantitative and qualitative serological assays. However, experimental challenge of the experimentally immunized horses by our standard virulent EIAV(PV) strain by using a low-dose multiple exposure protocol (three inoculations with 10 median horse infective doses, administered intravenously) revealed a marked difference in the protective efficacy of the various attenuated proviral vaccine strains that was evidently associated with the extent of vaccine virus attenuation, time of viral challenge, and the apparent maturation of virus-specific immunity.
Collapse
Affiliation(s)
- Jodi K Craigo
- Department of Molecular Genetics and Biochemistry, W1144 Biomedical Science Tower, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Luo K, Liu B, Xiao Z, Yu Y, Yu X, Gorelick R, Yu XF. Amino-terminal region of the human immunodeficiency virus type 1 nucleocapsid is required for human APOBEC3G packaging. J Virol 2004; 78:11841-52. [PMID: 15479826 PMCID: PMC523292 DOI: 10.1128/jvi.78.21.11841-11852.2004] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
APOBEC3G exerts its antiviral activity by targeting to retroviral particles and inducing viral DNA hypermutations in the absence of Vif. However, the mechanism by which APOBEC3G is packaged into virions remains unclear. We now report that viral genomic RNA enhances but is not essential for human APOBEC3G packaging into human immunodeficiency virus type 1 (HIV-1) virions. Packaging of APOBEC3G was also detected in HIV-1 Gag virus-like particles (VLP) that lacked all the viral genomic RNA packaging signals. Human APOBEC3G could be packaged efficiently into a divergent subtype HIV-1, as well as simian immunodeficiency virus, strain mac, and murine leukemia virus Gag VLP. Cosedimentation of human APOBEC3G and intracellular Gag complexes was detected by equilibrium density and velocity sucrose gradient analysis. Interaction between human APOBEC3G and HIV-1 Gag was also detected by coimmunoprecipitation experiments. This interaction did not require p6, p1, or the C-terminal region of NCp7. However, the N-terminal region, especially the first 11 amino acids, of HIV-1 NCp7 was critical for HIV-1 Gag and APOBEC3G interaction and virion packaging. The linker region flanked by the two active sites of human APOBEC3G was also important for efficient packaging into HIV-1 Gag VLP. Association of human APOBEC3G with RNA-containing intracellular complexes was observed. These results suggest that the N-terminal region of HIV-1 NC, which is critical for binding to RNA and mediating Gag-Gag oligomerization, plays an important role in APOBEC3G binding and virion packaging.
Collapse
Affiliation(s)
- Kun Luo
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Kristbjörnsdóttir HB, Andrésdóttir V, Svansson V, Torsteinsdóttir S, Matthíasdóttir S, Andrésson OS. The vif gene of maedi-visna virus is essential for infectivity in vivo and in vitro. Virology 2004; 318:350-9. [PMID: 14972560 DOI: 10.1016/j.virol.2003.09.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Revised: 09/25/2003] [Accepted: 09/29/2003] [Indexed: 11/17/2022]
Abstract
We have investigated the role of vif in maedi-visna virus (MVV), a lentivirus of sheep, by studying in vitro replication of vif-deleted MVV in several cell types, and the effects of vif deletion on in vivo infection. By measuring RT activity, we found that in comparison to wild-type MVV, growth of vif-deleted MVV was similar in fetal ovine synovial (FOS) cells, highly attenuated in sheep choroid plexus (SCP) cells, and not detectable in macrophages, natural target cells of MVV. Productive infection by vif-deleted MVV could not be demonstrated in sheep. An increased mutation frequency was observed in DNA produced by endogenous reverse transcription of viral RNA in vif-deleted virions, indicating the existence of a factor comparable in action to human APOBEC3G. These results suggest that the vif gene of MVV is essential for infectivity and that the Vif protein protects the viral genome from enpackaged mutagenic activities.
Collapse
|
25
|
Liu B, Yu X, Luo K, Yu Y, Yu XF. Influence of primate lentiviral Vif and proteasome inhibitors on human immunodeficiency virus type 1 virion packaging of APOBEC3G. J Virol 2004; 78:2072-81. [PMID: 14747572 PMCID: PMC369424 DOI: 10.1128/jvi.78.4.2072-2081.2004] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Vif protein of human immunodeficiency virus type 1 (HIV-1) is essential for viral evasion of the host antiviral protein APOBEC3G, also known as CEM15. Vif mutant but not wild-type HIV-1 viruses produced in the presence of APOBEC3G have been shown to undergo hypermutations in newly synthesized viral DNA upon infection of target cells, presumably resulting from C-to-U modification during minus-strand viral DNA synthesis. We now report that HIV-1 Vif could induce rapid degradation of human APOBEC3G that was blocked by the proteasome inhibitor MG132. The efficiency of Vif-induced downregulation of APOBEC3G expression depended on the level of Vif expression. A single amino acid substitution in the conserved SLQXLA motif reduced Vif function. Vif proteins from distantly related primate lentiviruses such as SIVagm were unable to suppress the antiviral activity of human APOBEC3G or the packaging of APOBEC3G into HIV-1 Vif mutant virions, due to a lack of interaction with human APOBEC3G. In the presence of the proteasome inhibitor MG132, virion-associated Vif increased dramatically. However, increased virion packaging of Vif did not prevent virion packaging of APOBEC3G when proteasome function was impaired, and the infectivity of these virions was significantly reduced. These results suggest that Vif function is required during virus assembly to remove APOBEC3G from packaging into released virions. Once packaged, virion-associated Vif could not efficiently block the antiviral activity of APOBEC3G.
Collapse
Affiliation(s)
- Bindong Liu
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
26
|
Pistello M, Matteucci D, Bonci F, Isola P, Mazzetti P, Zaccaro L, Merico A, Del Mauro D, Flynn N, Bendinelli M. AIDS vaccination studies using an ex vivo feline immunodeficiency virus model: protection from an intraclade challenge administered systemically or mucosally by an attenuated vaccine. J Virol 2003; 77:10740-50. [PMID: 14512524 PMCID: PMC224962 DOI: 10.1128/jvi.77.20.10740-10750.2003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Feline immunodeficiency virus (FIV) infection of domestic cats represents a valuable system through which to investigate criteria for antilentiviral vaccines in a natural host species. Here, we examined whether vaccination with a strain of FIV attenuated as a result of prolonged growth in vitro could protect against a fully virulent, highly heterologous intraclade challenge. The results indicated that the vaccine virus produced a low-grade infection with no detectable pathological effects and afforded a long-lasting sterilizing immunity if the challenge was delivered intraperitoneally as cell-free virus but not against a cell-associated intravaginal challenge. In the latter case, however, the replication and pathological consequences of the challenge virus were markedly suppressed. Together with similar results obtained in rhesus monkey models, these findings should give impulse to the development of attenuated FIV vaccines to be tested in controlled studies in field cats. Field studies may provide answers to some of the existing safety concerns surrounding attenuated AIDS vaccines in humans.
Collapse
Affiliation(s)
- Mauro Pistello
- Retrovirus Center and Virology Section, Department of Experimental Pathology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pedersen NC, North TW, Rigg R, Reading C, Higgins J, Leutenegger C, Henderson GL. 16alpha-Bromo-epiandrosterone therapy modulates experimental feline immunodeficiency virus viremia: initial enhancement leading to long-term suppression. Vet Immunol Immunopathol 2003; 94:133-48. [PMID: 12909410 DOI: 10.1016/s0165-2427(03)00081-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
16alpha-Bromo-epiandrosterone (epiBr), a synthetic derivative of the natural hormone dehyroepiandrosterone (DHEA), was evaluated for its effects on feline immunodeficiency virus (FIV) infection in experimental cats. The rationale for this study was based on the ability of DHEA to significantly reduce the mortality to viral infections in mice. DHEA and epiBr also have demonstrable in vitro anti-viral activity for both HIV-1 and FIV. Preliminary pharmacokinetic studies in cats demonstrated that subcutaneously injected epiBr was rapidly absorbed, completely metabolized, and nontoxic. Metabolites were excreted in both urine and feces, with the latter having the most complex pattern of breakdown products. Cats were then divided into four groups; two groups were infected with FIV and two uninfected. Two groups, one infected and one uninfected were treated on 5 consecutive days of weeks 0, 4, 8, 12 and 16 with epiBr. The remaining two groups were mock treated with the drug vehicle alone. Treatment started 1 week prior to infection and extended for 4 weeks after infection. Cats were observed for 20 weeks post-FIV infection. Infected cats had identical decreases in blood neutrophil and lymphocyte counts following, regardless of whether they were treated with epiBr or vehicle alone. The CD4/CD8 T-cell ratio was decreased following FIV exposure, but was significantly more decreased for the epiBr treated animals from week 2 post-infection onward. CD4+ T cells were decreased in FIV-infected cats treated with epiBr compared to their untreated cohort, while CD8+ T cells tended to be higher in treated animals. FIV infected cats that were treated with epiBr had over one-log higher virus loads at week 2 post-infection than non-epiBr treated cohorts. In spite of this enhanced initial viremia, the subsequent levels of virus in the blood were significantly lower in epiBr treated versus untreated animals. EpiBr treated cats had significantly higher FIV-p24 antibody responses than control cats receiving vehicle alone, although primary and secondary antibody responses to a T-cell dependent non-FIV antigen, keyhole limpet hemocyanin (KLH), were unaffected. EpiBr treatment significantly decreased the expected FIV-induced suppression of IL-12 p40 mRNA levels in peripheral blood mononuclear cells (PBMCs) observed at weeks 4, 5, 8, 9 and 16 post-infection, but had no influence on FIV-induced changes in IL-4, IL-6, IL-10, IFN-gamma, MIP-1alpha and RANTES.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacokinetics
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/urine
- Androsterone/analogs & derivatives
- Androsterone/pharmacokinetics
- Androsterone/pharmacology
- Androsterone/urine
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/blood
- CD4-CD8 Ratio/veterinary
- Cats
- Cytokines/biosynthesis
- Cytokines/blood
- Cytokines/genetics
- Enzyme-Linked Immunosorbent Assay/veterinary
- Feces/chemistry
- Feline Acquired Immunodeficiency Syndrome/drug therapy
- Feline Acquired Immunodeficiency Syndrome/immunology
- Feline Acquired Immunodeficiency Syndrome/metabolism
- Feline Acquired Immunodeficiency Syndrome/virology
- Immunodeficiency Virus, Feline/genetics
- Immunodeficiency Virus, Feline/immunology
- Immunodeficiency Virus, Feline/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Male
- Polymerase Chain Reaction/veterinary
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Random Allocation
- Specific Pathogen-Free Organisms
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Viremia/drug therapy
- Viremia/immunology
- Viremia/veterinary
Collapse
Affiliation(s)
- Niels C Pedersen
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Busch M, Lu D, Fritts L, Lifson JD, Miller CJ. Comparison of virology and immunology in SHIV 89.6 proviral DNA and virus-inoculated rhesus macaques. J Med Primatol 2003; 32:240-6. [PMID: 14498984 DOI: 10.1034/j.1600-0684.2003.00029.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Inoculation of cats, goats and monkeys with plasmids encoding full-length proviral genomes results in persistent lentiviral infections. This system could be used as a method for administration of an attenuated human immunodeficiency virus (HIV) vaccine. Here, we compare the virology and immunology in rhesus macaques inoculated with either simian/human immunodeficiency virus 89.6 (SHIV 89.6) virus or a plasmid containing the SHIV 89.6 proviral genome. There was a delay in appearance of systemic infection in DNA-inoculated animals compared with virus-inoculated animals, but otherwise the pattern of infection was similar. The serum immunoglobulin G anti-simian immunodeficiency virus (SIV) binding antibody response in DNA-inoculated animals was also delayed compared with virus-inoculated animals, but ultimately there was no difference between live virus and DNA-inoculation in the ability to induce the anti-SIV immune responses that were measured. Thus, the data support the concept that plasmid DNA encoding an attenuated virus could be used instead live virus for vaccination.
Collapse
Affiliation(s)
- Marc Busch
- California National Primate Research Center and Center for Comparative Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
29
|
Steinrigl A, Klein D. Phylogenetic analysis of feline immunodeficiency virus in Central Europe: a prerequisite for vaccination and molecular diagnostics. J Gen Virol 2003; 84:1301-1307. [PMID: 12692297 DOI: 10.1099/vir.0.18736-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Feline immunodeficiency virus (FIV) is a worldwide-occurring lentivirus that severely impairs the immune function of infected domestic cats. Due to structural and biological similarities, FIV represents a promising model for human immunodeficiency virus (HIV) and AIDS. A major obstacle in developing vaccines against lentiviruses is their high mutation rate. Furthermore, mutations in target sequences provide a pitfall for molecular diagnostics. It is therefore important to determine the genetic diversity of lentiviruses in any region where vaccination or implementation of new diagnostic techniques are planned. This study presents a phylogenetic analysis of 30 FIV strains derived from Central Europe. In order to improve the reliability of genotyping, DNA from two different proviral genes was amplified and comparative phylogenetic trees were inferred. The highly coincident results point to the existence of extensive virus variation with the presence of at least two highly divergent subtypes of FIV in Austria and Germany.
Collapse
Affiliation(s)
- Adolf Steinrigl
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Dieter Klein
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, A-1210 Vienna, Austria
| |
Collapse
|
30
|
Gaddis NC, Chertova E, Sheehy AM, Henderson LE, Malim MH. Comprehensive investigation of the molecular defect in vif-deficient human immunodeficiency virus type 1 virions. J Virol 2003; 77:5810-20. [PMID: 12719574 PMCID: PMC154025 DOI: 10.1128/jvi.77.10.5810-5820.2003] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Replication of human immunodeficiency virus type 1 (HIV-1) in primary blood lymphocytes, certain T-cell lines (nonpermissive cells), and most likely in vivo is highly dependent on the virally encoded Vif protein. Evidence suggests that Vif acts late in the viral life cycle during assembly, budding, and/or maturation to counteract the antiviral activity of the CEM15 protein and possibly other antiviral factors. Because HIV-1 virions produced in the absence of Vif are severely restricted at a postentry, preintegration step of infection, it is presumed that such virions differ from wild-type virions in some way. In the present study, we established a protocol for producing large quantities of vif-deficient HIV-1 (HIV-1/Delta vif) from an acute infection of nonpermissive T cells and performed a thorough examination of the defect in these virions. Aside from the expected lack of Vif, we observed no apparent abnormalities in the packaging, modification, processing, or function of proteins in Delta vif virions. In addition, we found no consistent defect in the ability of Delta vif virions to perform intravirion reverse transcription under a variety of assay conditions, suggesting that the reverse transcription complexes in these particles can behave normally under cell-free conditions. Consistent with this finding, neither the placement of the primer tRNA3Lys nor its ability to promote reverse transcription in an in vitro assay was affected by a lack of Vif. Based on the inability of this comprehensive analysis to uncover molecular defects in Delta vif virions, we speculate that such defects are likely to be subtle and/or rare.
Collapse
Affiliation(s)
- Nathan C Gaddis
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
31
|
Uhl E, Heaton-Jones T, Pu R, Yamamoto J. FIV vaccine development and its importance to veterinary and human medicine: a review FIV vaccine 2002 update and review. Vet Immunol Immunopathol 2002; 90:113-32. [PMID: 12459160 PMCID: PMC7119750 DOI: 10.1016/s0165-2427(02)00227-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2002] [Revised: 08/05/2002] [Accepted: 08/14/2002] [Indexed: 10/27/2022]
Abstract
Feline immunodeficiency virus (FIV) is a natural infection of domestic cats that results in acquired immunodeficiency syndrome resembling human immunodeficiency virus (HIV) infection in humans. The worldwide prevalence of FIV infection in domestic cats has been reported to range from 1 to 28%. Hence, an effective FIV vaccine will have an important impact on veterinary medicine in addition to being used as a small animal AIDS model for humans. Since the discovery of FIV reported in 1987, FIV vaccine research has pursued both molecular and conventional vaccine approaches toward the development of a commercial product. Published FIV vaccine trial results from 1998 to the present have been compiled to update the veterinary clinical and research communities on the immunologic and experimental efficacy status of these vaccines. A brief report is included on the outcome of the 10 years of collaborative work between industry and academia which led to recent USDA approval of the first animal lentivirus vaccine, the dual-subtype FIV vaccine. The immunogenicity and efficacy of the experimental prototype, dual-subtype FIV vaccine and the efficacy of the currently approved commercial, dual-subtype FIV vaccine (Fel-O-Vax FIV) are discussed. Potential cross-reactivity complications between commercial FIV diagnostic tests, Idexx Snap Combo Test and Western blot assays, and sera from previously vaccinated cats are also discussed. Finally, recommendations are made for unbiased critical testing of new FIV vaccines, the currently USDA approved vaccine, and future vaccines in development.
Collapse
Affiliation(s)
- E.W Uhl
- Department of Pathobiology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - T.G Heaton-Jones
- Department of Pathobiology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - R Pu
- Department of Pathobiology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - J.K Yamamoto
- Department of Pathobiology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| |
Collapse
|
32
|
Pistello M, Moscardini M, Mazzetti P, Bonci F, Zaccaro L, Isola P, Freer G, Specter S, Matteucci D, Bendinelli M. Development of feline immunodeficiency virus ORF-A (tat) mutants: in vitro and in vivo characterization. Virology 2002; 298:84-95. [PMID: 12093176 DOI: 10.1006/viro.2002.1442] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A functional ORF-A is essential for efficient feline immunodeficiency virus replication in lymphocytes. We have characterized a series of mutants of the Petaluma strain, derived from p34TF10 and having different combinations of stop codons and increasingly long deletions in ORF-A. Six clones proved fully replicative in fibroblastoid Crandell feline kidney cells and monocyte-derived macrophage cultures but failed to replicate in T cell lines and primary lymphoblasts. Cats inoculated with three selected mutants had considerably milder infections than controls given intact ORF-A virus. In vivo, the mutants maintained growth properties similar to those in vitro for at least 7 months, except that replication in lymphoid cells was strongly reduced but not ablated. One mutant underwent extensive ORF-A changes without, however, reverting to wild-type. Antiviral immune responses were feeble in all cats, suggesting that viral loads were too low to represent a sufficiently powerful antigenic stimulus.
Collapse
Affiliation(s)
- M Pistello
- Retrovirus Center and Virology Section, University of Pisa, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- M A Curran
- Stanford University, Department of Molecular Pharmacology, 269 Campus Drive, CCSR 3205A, Stanford, CA 94305-5174, USA
| | | |
Collapse
|
34
|
Pu R, Coleman J, Omori M, Arai M, Hohdatsu T, Huang C, Tanabe T, Yamamoto JK. Dual-subtype FIV vaccine protects cats against in vivo swarms of both homologous and heterologous subtype FIV isolates. AIDS 2001; 15:1225-37. [PMID: 11426067 DOI: 10.1097/00002030-200107060-00004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the immunogenicity and efficacy of an inactivated dual-subtype feline immunodeficiency virus (FIV) vaccine. DESIGN Specific-pathogen-free cats were immunized with dual-subtype (subtype A FIV(Pet) and subtype D FIV(Shi)) vaccine and challenged with either in vivo- or in vitro-derived FIV inocula. METHODS Dual-subtype vaccinated, single-subtype vaccinated, and placebo-immunized cats were challenged within vivo-derived heterologous subtype B FIV(Bang) [10--100 50% cat infectious doses (CID(50))], in vivo-derived homologous FIV(Shi)(50 CID(50)), and in vitro- and in vivo-derived homologous FIV(Pet)(20--50 CID(50)). Dual-subtype vaccine immunogenicity and efficacy were evaluated and compared to single-subtype strain vaccines. FIV infection was determined using virus isolation and proviral PCR of peripheral blood mononuclear cells and lymphoid tissues. RESULTS Four out of five dual-subtype vaccinated cats were protected against low-dose FIV(Bang) (10 CID(50)) and subsequently against in vivo-derived FIV(Pet) (50 CID(50)) challenge, whereas all placebo-immunized cats became infected. Furthermore, dual-subtype vaccine protected two out of five cats against high-dose FIV(Bang) challenge (100 CID(50)) which infected seven out of eight single-subtype vaccinated cats. All dual-subtype vaccinated cats were protected against in vivo-derived FIV(Pet), but only one out of five single-subtype vaccinated cats were protected against in vivo-derived FIV(Pet). Dual-subtype vaccination induced broad-spectrum virus-neutralizing antibodies and FIV-specific interferon-gamma responses along with elevated FIV-specific perforin mRNA levels, suggesting an increase in cytotoxic cell activities. CONCLUSION Dual-subtype vaccinated cats developed broad-spectrum humoral and cellular immunity which protected cats against in vivo-derived inocula of homologous and heterologous FIV subtypes. Thus, multi-subtype antigen vaccines may be an effective strategy against AIDS viruses.
Collapse
Affiliation(s)
- R Pu
- Department of Pathobiology, College of Veterinary Medicine, University of Florida, Gainesville, 32611-0880, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Mills J, Desrosiers R, Rud E, Almond N. Live attenuated HIV vaccines: a proposal for further research and development. AIDS Res Hum Retroviruses 2000; 16:1453-61. [PMID: 11054258 DOI: 10.1089/088922200750005976] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- J Mills
- Macfarlane Burnet Centre for Medical Research, Fairfield (Melbourne), Victoria, Australia.
| | | | | | | |
Collapse
|