1
|
Matsuura K, Yamaura M, Sakawaki H, Himeno A, Pisil Y, Kobayakawa T, Tsuji K, Tamamura H, Matsushita S, Miura T. Sensitivity to a CD4 mimic of a consensus clone of monkey-adapted CCR5-tropic SHIV-MK38C. Virology 2023; 578:171-179. [PMID: 36580864 DOI: 10.1016/j.virol.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
By acclimatizing CCR5-tropic tier 1B SHIV-MK1 to rhesus monkeys, a tier 2 SHIV-MK38 strain with neutralization resistance and high replication ability was generated. In this study, we generated SHIV-MK38C, a monkey-infectious consensus molecular clone of SHIV-MK38. Analysis using pseudotype viruses showed that MK38C was tier 1C because it lacked the N169D mutation, which is the most important mutation for neutralization resistance. MK38C harboring the N169D mutation became tier 2. However, the replication ability of SHIV-MK38C with N169D was low; more than 17 weeks elapsed before its detection in monkeys. Tier 1C MK38C was sensitive to a CD4 mimic. Therefore, SHIV-MK38C could be used to evaluate CD4 mimics in vivo.
Collapse
Affiliation(s)
- Kanako Matsuura
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mizuki Yamaura
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiromi Sakawaki
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ai Himeno
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yalcin Pisil
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takuya Kobayakawa
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Kohei Tsuji
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Hirokazu Tamamura
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Shuzo Matsushita
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Tomoyuki Miura
- Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
2
|
Pisil Y, Yazici Z, Shida H, Matsushita S, Miura T. Specific Substitutions in Region V2 of gp120 env confer SHIV Neutralisation Resistance. Pathogens 2020; 9:pathogens9030181. [PMID: 32138199 PMCID: PMC7157653 DOI: 10.3390/pathogens9030181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/16/2022] Open
Abstract
A tier 2 SHIV-MK38 strain was obtained after two in vivo passages of tier 1 SHIV-MK1. SHIV-MK38#818, cloned from the MK38 strain, was neutralisation-resistant, like the parental MK38 strain, to SHIV-infected monkey plasma (MP), HIV-1-infected human pooled plasma (HPP), and KD247 monoclonal antibody (mAb) (anti-V3 gp120 env). We investigated the mechanisms underlying the resistance of #818, specifically the amino acid substitutions that confer resistance to MK1. We introduced amino acid substitutions in the MK1 envelope by in vitro mutagenesis and then compared the neutralisation resistance to MP, HPP, and KD247 mAb with #818 in a neutralisation assay using TZM-bl cells. We selected 11 substitutions in the V1, V2, C2, V4, C4, and V5 regions based on the alignment of env of MK1 and #818. The neutralisation resistance of the mutant MK1s with 7 of 11 substitutions in the V1, C2, C4, and V5 regions did not change significantly. These substitutions did not alter any negative charges or N-glycans. The substitutions N169D and K187E, which added negative charges, and S190N in the V2 region of gp120 and A389T in V4, which created sites for N-glycan, conferred high neutralisation resistance. The combinations N169D+K187E, N169D+S190N, and N169D+A389T resulted in MK1 neutralisation resistance close to that of #818. The combinations without 169D were neutralisation-sensitive. Therefore, N169D is the most important substitution for neutralisation resistance. This study demonstrated that although the V3 region sequences of #818 and MK1 are the same, V3 binding antibodies cannot neutralise #818 pseudovirus. Instead, mutations in the V2 and V4 regions inhibit the neutralisation of anti-V3 antibodies. We hypothesised that 169D and 190N altered the MK1 Env conformation so that the V3 region is buried. Therefore, the V2 region may block KD247 from binding to the tip of the V3 region.
Collapse
Affiliation(s)
- Yalcin Pisil
- Laboratory of Primate Model, Research Center for Infectious Diseases, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto 615-8530, Japan;
| | - Zafer Yazici
- Department of Virology, Faculty of Veterinary Medicine, 19 Mayis University, Samsun 55270, Turkey;
| | - Hisatoshi Shida
- Division of Molecular Virology, Institute of Immunological Science, Hokkaido University, Hokkaido 060-0808, Japan;
| | - Shuzo Matsushita
- Center for AIDS Research, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Tomoyuki Miura
- Laboratory of Primate Model, Research Center for Infectious Diseases, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto 615-8530, Japan;
- Correspondence:
| |
Collapse
|
3
|
Iwanami S, Kakizoe Y, Morita S, Miura T, Nakaoka S, Iwami S. A highly pathogenic simian/human immunodeficiency virus effectively produces infectious virions compared with a less pathogenic virus in cell culture. Theor Biol Med Model 2017; 14:9. [PMID: 28431573 PMCID: PMC5401468 DOI: 10.1186/s12976-017-0055-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/18/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The host range of human immunodeficiency virus (HIV) is quite narrow. Therefore, analyzing HIV-1 pathogenesis in vivo has been limited owing to lack of appropriate animal model systems. To overcome this, chimeric simian and human immunodeficiency viruses (SHIVs) that encode HIV-1 Env and are infectious to macaques have been developed and used to investigate the pathogenicity of HIV-1 in vivo. So far, we have many SHIV strains that show different pathogenesis in macaque experiments. However, dynamic aspects of SHIV infection have not been well understood. To fully understand the dynamic properties of SHIVs, we focused on two representative strains-the highly pathogenic SHIV, SHIV-KS661, and the less pathogenic SHIV, SHIV-#64-and measured the time-course of experimental data in cell culture. METHODS We infected HSC-F with SHIV-KS661 and -#64 and measured the concentration of Nef-negative (target) and Nef-positive (infected) HSC-F cells, the total viral load, and the infectious viral load daily for 9 days. The experiments were repeated at two different multiplicities of infection, and a previously developed mathematical model incorporating the infectious and non-infectious viruses was fitted to the full dataset of each strain simultaneously to characterize the infection dynamics of these two strains. RESULTS AND CONCLUSIONS We quantified virological indices including virus burst sizes and basic reproduction number of both SHIV-KS661 and -#64. Comparing the burst size of total and infectious viruses (viral RNA copies and TCID50, respectively), we found that there was a statistically significant difference between the infectious virus burst size of SHIV-KS661 and -#64, while there was no significant difference between the total virus burst size. Furthermore, our analyses showed that the fraction of infectious virus among the produced SHIV-KS661 viruses, which is defined as the infectious viral load (TCID50/ml) divided by the total viral load (RNA copies/ml), is more than 10-fold higher than that of SHIV-#64 during overall infection (i.e., for 9 days). Taken together, we conclude that the highly pathogenic SHIV produces infectious virions more effectively than the less pathogenic SHIV in cell culture.
Collapse
Affiliation(s)
- Shoya Iwanami
- Department of Biology, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Yusuke Kakizoe
- Department of Biology, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Satoru Morita
- Department of Mathematical and Systems Engineering, Shizuoka University, Hamamatsu, Shizuoka, Japan
| | - Tomoyuki Miura
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shinji Nakaoka
- PRESTO, JST, Kawaguchi, Saitama, Japan.,Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Shingo Iwami
- Department of Biology, Kyushu University, Nishi-ku, Fukuoka, Japan. .,PRESTO, JST, Kawaguchi, Saitama, Japan. .,CREST, JST, Kawaguchi, Saitama, Japan.
| |
Collapse
|
4
|
Nonhuman Primate Models for Studies of AIDS Virus Persistence During Suppressive Combination Antiretroviral Therapy. Curr Top Microbiol Immunol 2017; 417:69-109. [PMID: 29026923 DOI: 10.1007/82_2017_73] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Nonhuman primate (NHP) models of AIDS represent a potentially powerful component of the effort to understand in vivo sources of AIDS virus that persist in the setting of suppressive combination antiretroviral therapy (cART) and to develop and evaluate novel strategies for more definitive treatment of HIV infection (i.e., viral eradication "cure", or sustained off-cART remission). Multiple different NHP models are available, each characterized by a particular NHP species, infecting virus, and cART regimen, and each with a distinct capacity to recapitulate different aspects of HIV infection. Given these different biological characteristics, and their associated strengths and limitations, different models may be preferred to address different questions pertaining to virus persistence and cure research, or to evaluate different candidate intervention approaches. Recent developments in improved cART regimens for use in NHPs, new viruses, a wider array of sensitive virologic assay approaches, and a better understanding of pathogenesis should allow even greater contributions from NHP models to this important area of HIV research in the future.
Collapse
|
5
|
Del Prete GQ, Lifson JD, Keele BF. Nonhuman primate models for the evaluation of HIV-1 preventive vaccine strategies: model parameter considerations and consequences. Curr Opin HIV AIDS 2016; 11:546-554. [PMID: 27559710 PMCID: PMC5100008 DOI: 10.1097/coh.0000000000000311] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Nonhuman primate (NHP) models of AIDS are powerful systems for evaluating HIV vaccine approaches in vivo. Authentic features of HIV-1 transmission, dissemination, target cell tropism, and pathogenesis, and aspects of anti-HIV-1 immune responses, can be recapitulated in NHPs provided the appropriate, specific model parameters are considered. Here, we discuss key model parameter options and their implications for HIV-1 vaccine evaluation. RECENT FINDINGS With the availability of several different NHP host species/subspecies, different challenge viruses and challenge stock production methods, and various challenge routes and schemata, multiple NHP models of AIDS exist for HIV vaccine evaluation. The recent development of multiple new challenge viruses, including chimeric simian-human immunodeficiency viruses and simian immunodeficiency virus clones, improved characterization of challenge stocks and production methods, and increased insight into specific challenge parameters have resulted in an increase in the number of available models and a better understanding of the implications of specific study design choices. SUMMARY Recent progress and technical developments promise new insights into basic disease mechanisms and improved models for better preclinical evaluation of interventions to prevent HIV transmission.
Collapse
Affiliation(s)
- Gregory Q. Del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| |
Collapse
|
6
|
Ishida Y, Yoneda M, Otsuki H, Watanabe Y, Kato F, Matsuura K, Kikukawa M, Matsushita S, Hishiki T, Igarashi T, Miura T. Generation of a neutralization-resistant CCR5 tropic simian/human immunodeficiency virus (SHIV-MK38) molecular clone, a derivative of SHIV-89.6. J Gen Virol 2016; 97:1249-1260. [PMID: 26850058 DOI: 10.1099/jgv.0.000421] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Previously, we reported that a new genetically diverse CCR5 (R5) tropic simian/human immunodeficiency virus (SHIV-MK38) adapted to rhesus monkeys became more neutralization resistant to SHIV-infected plasma than did the parental SHIV-KS661 clone. Here, to clarify the significance of the neutralization-resistant phenotype of SHIV in a macaque model, we initially investigated the precise neutralization phenotype of the SHIVs, including SHIV-MK38 molecular clones, using SHIV-MK38-infected plasma, a pooled plasma of human immunodeficiency virus (HIV)-infected individuals, soluble CD4 and anti-HIV-1 neutralizing mAbs, the epitopes of which were known. The results show that SHIV-KS661 had tier 1 neutralization sensitivity, but monkey-adapted R5 tropic SHIV-MK38 acquired neutralization resistance similar to that of tier 2 or 3 as a clone virus. Sequence analysis of the env gene suggested that the neutralization-resistant phenotype of SHIV-MK38 was acquired by conformational changes in Env associated with the net charge and potential N-linked glycosylation sites. To examine the relationship between neutralization phenotype and stably persistent infection in monkeys, we performed in vivo rectal inoculation experiments using a SHIV-MK38 molecular clone. The results showed that one of three rhesus monkeys exhibited durable infection with a plasma viral load of 105 copies ml- 1 despite the high antibody responses that occurred in the host. Whilst further improvements are required in the development of a challenge virus, it will be useful to generate a neutralization-resistant R5 tropic molecular clone of the SHIV-89.6 lineage commonly used for vaccine development - a result that can be used to explore the foundation of AIDS pathogenesis.
Collapse
Affiliation(s)
- Yuki Ishida
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Mai Yoneda
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Hiroyuki Otsuki
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Yuji Watanabe
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Fumihiro Kato
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Kanako Matsuura
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Minako Kikukawa
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Shuzo Matsushita
- Division of Clinical Retrovirology and Infectious Diseases, Center for AIDS Research,Kumamoto University, Kumamoto 860-0811,Japan
| | - Takayuki Hishiki
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Tatsuhiko Igarashi
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| | - Tomoyuki Miura
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research,Kyoto University, 53 Shogoinkawaharacho, Sakyo-ku, Kyoto 606-8507,Japan
| |
Collapse
|
7
|
Kakizoe Y, Nakaoka S, Beauchemin CAA, Morita S, Mori H, Igarashi T, Aihara K, Miura T, Iwami S. A method to determine the duration of the eclipse phase for in vitro infection with a highly pathogenic SHIV strain. Sci Rep 2015; 5:10371. [PMID: 25996439 PMCID: PMC4440524 DOI: 10.1038/srep10371] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/09/2015] [Indexed: 01/08/2023] Open
Abstract
The time elapsed between successful cell infection and the start of virus production is called the eclipse phase. Its duration is specific to each virus strain and, along with an effective virus production rate, plays a key role in infection kinetics. How the eclipse phase varies amongst cells infected with the same virus strain and therefore how best to mathematically represent its duration is not clear. Most mathematical models either neglect this phase or assume it is exponentially distributed, such that at least some if not all cells can produce virus immediately upon infection. Biologically, this is unrealistic (one must allow for the translation, transcription, export, etc. to take place), but could be appropriate if the duration of the eclipse phase is negligible on the time-scale of the infection. If it is not, however, ignoring this delay affects the accuracy of the mathematical model, its parameter estimates, and predictions. Here, we introduce a new approach, consisting in a carefully designed experiment and simple analytical expressions, to determine the duration and distribution of the eclipse phase in vitro. We find that the eclipse phase of SHIV-KS661 lasts on average one day and is consistent with an Erlang distribution.
Collapse
Affiliation(s)
- Yusuke Kakizoe
- Department of Biology, Kyushu University, Fukuoka 812-8581, Japan
| | - Shinji Nakaoka
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | | | - Satoru Morita
- Department of Mathematical and Systems Engineering, Shizuoka University, Shizuoka 432-8561, Japan
| | - Hiromi Mori
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | - Kazuyuki Aihara
- 1] Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan [2] Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8656, Japan
| | - Tomoyuki Miura
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Shingo Iwami
- 1] Department of Biology, Kyushu University, Fukuoka 812-8581, Japan [2] PRESTO, JST, Kawaguchi, Saitama 3320012, Japan [3] CREST, JST, Kawaguchi, Saitama 3320012, Japan
| |
Collapse
|
8
|
Kakizoe Y, Morita S, Nakaoka S, Takeuchi Y, Sato K, Miura T, Beauchemin CAA, Iwami S. A conservation law for virus infection kinetics in vitro. J Theor Biol 2015; 376:39-47. [PMID: 25882746 DOI: 10.1016/j.jtbi.2015.03.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/13/2015] [Accepted: 03/28/2015] [Indexed: 01/18/2023]
Abstract
Conservation laws are among the most important properties of a physical system, but are not commonplace in biology. We derived a conservation law from the basic model for viral infections which consists in a small set of ordinary differential equations. We challenged the conservation law experimentally for the case of a virus infection in a cell culture. We found that the derived, conserved quantity remained almost constant throughout the infection period, implying that the derived conservation law holds in this biological system. We also suggest a potential use for the conservation law in evaluating the accuracy of experimental measurements.
Collapse
Affiliation(s)
- Yusuke Kakizoe
- Department of Biology, Kyushu University, Fukuoka, Japan
| | - Satoru Morita
- Department of Mathematical and Systems Engineering, Shizuoka University, Shizuoka, Japan
| | - Shinji Nakaoka
- Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yasuhiro Takeuchi
- College of Science and Engineering, Aoyama Gakuin University, Kanagawa, Japan
| | - Kei Sato
- Institute for Virus Research, Kyoto University, Kyoto, Japan; CREST, JST, Saitama, Japan
| | - Tomoyuki Miura
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | - Shingo Iwami
- Department of Biology, Kyushu University, Fukuoka, Japan; CREST, JST, Saitama, Japan; PRESTO, JST, Saitama, Japan.
| |
Collapse
|
9
|
A critical analysis of the cynomolgus macaque, Macaca fascicularis, as a model to test HIV-1/SIV vaccine efficacy. Vaccine 2014; 33:3073-83. [PMID: 25510387 DOI: 10.1016/j.vaccine.2014.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/26/2014] [Accepted: 12/03/2014] [Indexed: 02/07/2023]
Abstract
The use of a number of non-rhesus macaque species, but especially cynomolgus macaques as a model for HIV-1 vaccine development has increased in recent years. Cynomolgus macaques have been used in the United Kingdom, Europe, Canada and Australia as a model for HIV vaccine development for many years. Unlike rhesus macaques, cynomolgus macaques infected with SIV show a pattern of disease pathogenesis that more closely resembles that of human HIV-1 infection, exhibiting lower peak and set-point viral loads and slower progression to disease with more typical AIDS defining illnesses. Several advances have been made recently in the use of the cynomolgus macaque SIV challenge model that allow the demonstration of vaccine efficacy using attenuated viruses and vectors that are both viral and non-viral in origin. This review aims to probe the details of various vaccination trials carried out in cynomolgus macaques in the context of our modern understanding of the highly diverse immunogenetics of this species with a view to understanding the species-specific immune correlates of protection and the efficacy of vectors that have been used to design vaccines.
Collapse
|
10
|
Baroncelli S, Negri DRM, Michelini Z, Cara A. Macaca mulatta,fascicularisandnemestrinain AIDS vaccine development. Expert Rev Vaccines 2014; 7:1419-34. [DOI: 10.1586/14760584.7.9.1419] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Kaneyasu K, Kita M, Ohkura S, Yamamoto T, Ibuki K, Enose Y, Sato A, Kodama M, Miura T, Hayami M. Protective Efficacy of Nonpathogenic Nef-Deleted SHIV Vaccination Combined with Recombinant IFN-γ Administration against a Pathogenic SHIV Challenge in Rhesus Monkeys. Microbiol Immunol 2013; 49:1083-94. [PMID: 16365534 DOI: 10.1111/j.1348-0421.2005.tb03706.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We previously reported that a nef-deleted SHIV (SHIV-NI) is nonpathogenic and gave macaques protection from challenge infection with pathogenic SHIV-C2/1. To investigate whether IFN-gamma augments the immune response induced by this vaccination, we examined the antiviral and adjuvant effect of recombinant human IFN-gamma (rIFN-gamma) in vaccinated and unvaccinated monkeys. Nine monkeys were vaccinated with nef-deleted nonpathogenic SHIV-NI. Four of them were administered with rIFN-gamma and the other five monkeys were administered with placebo. After the challenge with pathogenic SHIV-C2/1, CD4(+) T-cell counts were maintained similarly in monkeys of both groups, while those of the unvaccinated monkeys decreased dramatically at 2 weeks after challenge. However, the peaks of plasma viral load were reduced to 100-fold in SHIV-NI vaccinated monkeys combined with rIFN-gamma compared with those in SHIV-NI vaccinated monkeys without rIFN-gamma. The peaks of plasma viral load were inversely correlated with the number of SIV Gag-specific IFN-gamma-producing cells. In SHIV-NI-vaccinated monkeys with rIFN-gamma, the number of SIV Gag-specific IFN-gamma-producing cells of PBMCs increased 2-fold compared with those in SHIV-NI-vaccinated monkeys without rIFN-gamma, and the NK activity and MIP-1alpha production of PBMCs were also enhanced. Thus, vaccination of SHIV-NI in combination with rIFN-gamma was more effective in modulating the antiviral immune system into a Th1 type response than SHIV-NI vaccination alone. These results suggest that IFN-gamma augmented the anti-viral effect by enhancing innate immunity and shifting the immune response to Th1.
Collapse
Affiliation(s)
- Kentaro Kaneyasu
- Institute for Virus Research, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Otsuki H, Hishiki T, Miura T, Hashimoto C, Narumi T, Tamamura H, Yoshimura K, Matsushita S, Igarashi T. Generation of a replication-competent simian-human immunodeficiency virus, the neutralization sensitivity of which can be enhanced in the presence of a small-molecule CD4 mimic. J Gen Virol 2013; 94:2710-2716. [PMID: 24026672 DOI: 10.1099/vir.0.055590-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Simian-human immunodeficiency virus (SHIV) carrying the envelope from the clade B clinical human immunodeficiency virus type 1 (HIV-1) isolate MNA, designated SHIV MNA, was generated through intracellular homologous recombination. SHIV MNA inherited biological properties from the parental HIV-1, including CCR5 co-receptor preference, resistance to neutralization by the anti-V3 loop mAb KD-247 and loss of resistance in the presence of the CD4-mimic small-molecule YYA-021. SHIV MNA showed productive replication in rhesus macaque PBMCs. Experimental infection of a rhesus macaque with SHIV MNA caused a transient but high titre of plasma viral RNA and a moderate antibody response. Immunoglobulin in the plasma at 24 weeks post-infection was capable of neutralizing SHIV MNA in the presence but not in the absence of YYA-021. SHIV MNA could serve a model for development of novel therapeutic interventions based on CD4-mimic-mediated conversion of envelope protein susceptible to antibody neutralization.
Collapse
Affiliation(s)
- Hiroyuki Otsuki
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Takayuki Hishiki
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Tomoyuki Miura
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Chie Hashimoto
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Tetsuo Narumi
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Hirokazu Tamamura
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Kazuhisa Yoshimura
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Shuzo Matsushita
- Division of Clinical Retrovirology and Infectious Diseases, Center for AIDS Research, Kumamoto University, Kumamoto 860-0811, Japan
| | - Tatsuhiko Igarashi
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
13
|
Matsuyama-Murata M, Inaba K, Horiuchi R, Fukazawa Y, Ibuki K, Hayami M, Miura T. Genetic similarity of circulating and small intestinal virus at the end stage of acute pathogenic simian-human immunodeficiency virus infection. Front Microbiol 2013; 4:204. [PMID: 23885255 PMCID: PMC3717482 DOI: 10.3389/fmicb.2013.00204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/01/2013] [Indexed: 11/13/2022] Open
Abstract
To understand the pathogenicity of acquired immune deficiency syndrome (AIDS), it is important to clarify where, when and how the virus replicates in the body of infected individuals. To identify the major virus replication site at the end stage of SHIV infection, we investigated the systemic tissues of SHIV-infected monkeys that developed AIDS-like disease. We quantified proviral DNA, and compared the mutation patterns of the viruses in various systemic tissues and in peripheral blood through phylogenetic analysis of the full genome sequence. We found that the amounts of proviral DNA detected in internal tissues were higher than those in peripheral blood mononuclear cells. In the sequence and phylogenetic tree analyses, the mutation patterns of the viruses in each tissue were generally different. However, the mutation pattern of the viruses in the jejunum and mesenteric lymph node were most similar to that of plasma viral RNA among the tissues examined in all three monkeys. In two of the three monkeys, which were euthanized earlier, viruses in the jejunum and mesenteric lymph node occupied the root position of the phylogenetic tree. Furthermore, in these tissues, more than 50% of SHIV-expressing cells were identified as macrophages based on co-expression of CD68. These results suggest that macrophages of the small intestine and/or mesenteric lymph node are the major virus production site at the end stage of SHIV infection of macaques.
Collapse
Affiliation(s)
- Megumi Matsuyama-Murata
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
14
|
Nakasone T, Murakami T, Yamamoto N. Double oral administration of emtricitabine/tenofovir prior to virus exposure protects against highly pathogenic simian/human immunodeficiency virus infection in macaques. Jpn J Infect Dis 2012; 65:345-9. [PMID: 22814162 DOI: 10.7883/yoken.65.345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the absence of any effective vaccine against human immunodeficiency virus (HIV), current anti-retroviral drugs may be suitable for pre-exposure prophylaxis (PrEP). Previous large clinical trials showed that PrEP reduced HIV infection in high-risk populations. Emtricitabine/tenofovir (FTC/TDF) may be a suitable agent for PrEP. FTC/TDF PrEP efficacy was evaluated using a highly pathogenic simian/human immunodeficiency virus (SHIV) in a non-human primate model of AIDS, the SHIV-KS661c/cynomolgus monkey model. Double oral administration of FTC/TDF (20/30 mg/kg), at 24 h and a few minutes prior to exposure, completely protected 2/3 monkeys from infection. Interestingly, a single oral administration 2 weeks before viral exposure moderately rescued CD4 cells, although the data did not reach statistical significance. These results are consistent with previous primate studies and with recent clinical data.
Collapse
Affiliation(s)
- Tadashi Nakasone
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan. nakabone@nih.go.jp
| | | | | |
Collapse
|
15
|
Fujita Y, Otsuki H, Watanabe Y, Yasui M, Kobayashi T, Miura T, Igarashi T. Generation of a replication-competent chimeric simian-human immunodeficiency virus carrying env from subtype C clinical isolate through intracellular homologous recombination. Virology 2012; 436:100-11. [PMID: 23219366 DOI: 10.1016/j.virol.2012.10.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 09/10/2012] [Accepted: 10/29/2012] [Indexed: 11/28/2022]
Abstract
A new simian-human immunodeficiency virus (SHIV), carrying env from an uncloned HIV-1 subtype C clinical isolate (97ZA012), was generated through intracellular homologous recombination, a DNA repair mechanism of the host cell. PCR fragments amplified from an existing SHIV plasmid (a 7-kb fragment from the 5' end and a 1.5-kb fragment from the 3' end) and a 4-kb fragment amplified from 97ZA012 cDNA containing env were co-transfected to human lymphoid cells. The resulting recombinant was subjected to serial passage in rhesus peripheral blood mononuclear cells (RhPBMCs). The resulting SHIV 97ZA012 was replication competent in RhPBMCs and monkey alveolar macrophages, and possessed CCR5 preference as an entry co-receptor. Experimental infection of rhesus macaques with SHIV 97ZA012 caused high titers of plasma viremia and a transient but profound depletion of CD4(+) T lymphocytes in the lung. Animal-to-animal passage was shown to be a promising measure for further adaptation of the virus in monkeys.
Collapse
Affiliation(s)
- Yasuhisa Fujita
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Iwami S, Sato K, De Boer RJ, Aihara K, Miura T, Koyanagi Y. Identifying viral parameters from in vitro cell cultures. Front Microbiol 2012; 3:319. [PMID: 22969758 PMCID: PMC3432869 DOI: 10.3389/fmicb.2012.00319] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Accepted: 08/16/2012] [Indexed: 11/13/2022] Open
Abstract
Current in vitro cell culture studies of viral replication deliver detailed time courses of several virological variables, like the amount of virions and the number of target cells, measured over several days of the experiment. Each of these time points solely provides a snap-shot of the virus infection kinetics and is brought about by the complex interplay of target cell infection, and viral production and cell death. It remains a challenge to interpret these data quantitatively and to reveal the kinetics of these underlying processes to understand how the viral infection depends on these kinetic properties. In order to decompose the kinetics of virus infection, we introduce a method to “quantitatively” describe the virus infection in in vitro cell cultures, and discuss the potential of the mathematical based analyses for experimental virology.
Collapse
Affiliation(s)
- Shingo Iwami
- Faculty of Sciences, Department of Biology, Kyushu University Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Single oral administration of the novel CXCR4 antagonist, KRH-3955, induces an efficient and long-lasting increase of white blood cell count in normal macaques, and prevents CD4 depletion in SHIV-infected macaques: a preliminary study. Med Microbiol Immunol 2012; 202:175-82. [DOI: 10.1007/s00430-012-0254-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/23/2012] [Indexed: 11/28/2022]
|
18
|
Iwami S, Holder BP, Beauchemin CAA, Morita S, Tada T, Sato K, Igarashi T, Miura T. Quantification system for the viral dynamics of a highly pathogenic simian/human immunodeficiency virus based on an in vitro experiment and a mathematical model. Retrovirology 2012; 9:18. [PMID: 22364292 PMCID: PMC3305505 DOI: 10.1186/1742-4690-9-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 02/25/2012] [Indexed: 01/15/2023] Open
Abstract
Background Developing a quantitative understanding of viral kinetics is useful for determining the pathogenesis and transmissibility of the virus, predicting the course of disease, and evaluating the effects of antiviral therapy. The availability of data in clinical, animal, and cell culture studies, however, has been quite limited. Many studies of virus infection kinetics have been based solely on measures of total or infectious virus count. Here, we introduce a new mathematical model which tracks both infectious and total viral load, as well as the fraction of infected and uninfected cells within a cell culture, and apply it to analyze time-course data of an SHIV infection in vitro. Results We infected HSC-F cells with SHIV-KS661 and measured the concentration of Nef-negative (target) and Nef-positive (infected) HSC-F cells, the total viral load, and the infectious viral load daily for nine days. The experiments were repeated at four different MOIs, and the model was fitted to the full dataset simultaneously. Our analysis allowed us to extract an infected cell half-life of 14.1 h, a half-life of SHIV-KS661 infectiousness of 17.9 h, a virus burst size of 22.1 thousand RNA copies or 0.19 TCID50, and a basic reproductive number of 62.8. Furthermore, we calculated that SHIV-KS661 virus-infected cells produce at least 1 infectious virion for every 350 virions produced. Conclusions Our method, combining in vitro experiments and a mathematical model, provides detailed quantitative insights into the kinetics of the SHIV infection which could be used to significantly improve the understanding of SHIV and HIV-1 pathogenesis. The method could also be applied to other viral infections and used to improve the in vitro determination of the effect and efficacy of antiviral compounds.
Collapse
Affiliation(s)
- Shingo Iwami
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Mycobacterium bovis Bacille Calmette-Guérin as a Vaccine Vector for Global Infectious Disease Control. Tuberc Res Treat 2011; 2011:574591. [PMID: 22567267 PMCID: PMC3335490 DOI: 10.1155/2011/574591] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/07/2011] [Indexed: 01/19/2023] Open
Abstract
Mycobacterium bovis bacille Calmette-Guérin (BCG) is the only available vaccine for tuberculosis (TB). Although this vaccine is effective in controlling infantile TB, BCG-induced protective effects against pulmonary diseases in adults have not been clearly demonstrated. Recombinant BCG (rBCG) technology has been extensively applied to obtain more potent immunogenicity of this vaccine, and several candidate TB vaccines have currently reached human clinical trials. On the other hand, recent progress in the improvement of the BCG vector, such as the codon optimization strategy and combination with viral vector boost, allows us to utilize this bacterium in HIV vaccine development. In this paper, we review recent progress in rBCG-based vaccine studies that may have implications in the development of novel vaccines for controlling global infectious diseases in the near future.
Collapse
|
20
|
Matsuda K, Inaba K, Fukazawa Y, Matsuyama M, Ibuki K, Horiike M, Saito N, Hayami M, Igarashi T, Miura T. In vivo analysis of a new R5 tropic SHIV generated from the highly pathogenic SHIV-KS661, a derivative of SHIV-89.6. Virology 2010; 399:134-143. [PMID: 20102777 DOI: 10.1016/j.virol.2010.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 12/14/2009] [Accepted: 01/05/2010] [Indexed: 10/19/2022]
Abstract
Although X4 tropic SHIVs have been studied extensively, they show distinct infection phenotypes from those of R5 tropic viruses, which play an important role in HIV-1 transmission and pathogenesis. To augment the variety of R5 tropic SHIVs, we generated a new R5 tropic SHIV from the highly pathogenic X4 tropic SHIV-KS661, a derivative of SHIV-89.6. Based on consensus amino acid alignment analyses of subtype B R5 tropic HIV-1, five amino acid substitutions in the third variable region successfully changed the secondary receptor preference from X4 to R5. Improvements in viral replication were observed in infected rhesus macaques after two passages, and reisolated virus was designated SHIV-MK38. SHIV-MK38 maintained R5 tropism through in vivo passages and showed robust replication in infected monkeys. Our study clearly demonstrates that a minimal number of amino acid substitutions in the V3 region can alter secondary receptor preference and increase the variety of R5 tropic SHIVs.
Collapse
Affiliation(s)
- Kenta Matsuda
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Katsuhisa Inaba
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshinori Fukazawa
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Megumi Matsuyama
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Ibuki
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mariko Horiike
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoki Saito
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanori Hayami
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuhiko Igarashi
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoyuki Miura
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawaramachi, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
21
|
Inaba K, Fukazawa Y, Matsuda K, Himeno A, Matsuyama M, Ibuki K, Miura Y, Koyanagi Y, Nakajima A, Blumberg RS, Takahashi H, Hayami M, Igarashi T, Miura T. Small intestine CD4+ cell reduction and enteropathy in simian/human immunodeficiency virus KS661-infected rhesus macaques in the presence of low viral load. J Gen Virol 2009; 91:773-81. [PMID: 19889928 DOI: 10.1099/vir.0.017368-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Human immunodeficiency virus type 1, simian immunodeficiency virus and simian/human immunodeficiency virus (SHIV) infection generally lead to death of the host accompanied by high viraemia and profound CD4(+) T-cell depletion. SHIV clone KS661-infected rhesus macaques with a high viral load set point (HVL) ultimately experience diarrhoea and wasting at 6-12 months after infection. In contrast, infected macaques with a low viral load set point (LVL) usually live asymptomatically throughout the observation period, and are therefore referred to as asymptomatic LVL (Asym LVL) macaques. Interestingly, some LVL macaques exhibit diarrhoea and wasting similar to the symptoms of HVL macaques and are termed symptomatic LVL (Sym LVL) macaques. This study tested the hypothesis that Sym LVL macaques have the same degree of intestinal abnormalities as HVL macaques. The proviral DNA loads in lymphoid tissue and the intestines of Sym LVL and Asym LVL macaques were comparable and all infected monkeys showed villous atrophy. Notably, the CD4(+) cell frequencies of lymphoid tissues and intestines in Sym LVL macaques were remarkably lower than those in Asym LVL and uninfected macaques. Furthermore, Sym LVL and HVL macaques exhibited an increased number of activated macrophages. In conclusion, intestinal disorders including CD4(+) cell reduction and abnormal immune activation can be observed in SHIV-KS661-infected macaques independent of virus replication levels.
Collapse
Affiliation(s)
- Katsuhisa Inaba
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Postinfection passive transfer of KD-247 protects against simian/human immunodeficiency virus-induced CD4+ T-cell loss in macaque lymphoid tissue. AIDS 2009; 23:1485-94. [PMID: 19528788 DOI: 10.1097/qad.0b013e32832e5331] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Preadministration of high-affinity humanized anti-HIV-1 mAb KD-247 by passive transfer provides sterile protection of monkeys from heterologous chimeric simian/human immunodeficiency virus infection. METHODS Beginning 1 h, 1 day, or 1 week after simian/human immunodeficiency virus-C2/1 challenge (20 50% tissue culture infective dose), mature, male cynomolgus monkeys received multiple passive transfers of KD-247 (45 mg/kg) on a weekly basis for approximately 2 months. Concentrations and viral loads were measured in peripheral blood, and CD4 T-cell counts were examined in both peripheral blood and various lymphoid tissues. RESULTS Pharmacokinetic examination revealed similar plasma maintenance levels ranging from 200 to 500 microg/ml of KD-247 in the three groups. One of the six monkeys given KD-247 could not maintain these concentrations, and elicitation of anti-KD-247 idiotype antibody was suggested. All monkeys given KD-247 exhibited striking postinfection protection against both CD4 T-cell loss in various lymphoid tissues and atrophic changes in organs compared with control group animals treated with normal human immunoglobulin G. The KD-247-treated groups were also partially protected against plasma viral load elevation in peripheral blood samples, although the complete protection previously reported with preadministration of this mAb was not achieved. CONCLUSION Postinfection passive transfer of humanized mAb KD-247 with strong neutralizing capacity against challenged virus simian/human immunodeficiency virus-C2/1 protected CD4 T cells in lymphoid organs.
Collapse
|
23
|
Small intestine CD4+ T cells are profoundly depleted during acute simian-human immunodeficiency virus infection, regardless of viral pathogenicity. J Virol 2008; 82:6039-44. [PMID: 18400862 DOI: 10.1128/jvi.02753-07] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To analyze the relationship between acute virus-induced injury and the subsequent disease phenotype, we compared the virus replication and CD4(+) T-cell profiles for monkeys infected with isogenic highly pathogenic (KS661) and moderately pathogenic (#64) simian-human immunodeficiency viruses (SHIVs). Intrarectal infusion of SHIV-KS661 resulted in rapid, systemic, and massive virus replication, while SHIV-#64 replicated more slowly and reached lower titers. Whereas KS661 systemically depleted CD4(+) T cells, #64 caused significant CD4(+) T-cell depletion only in the small intestine. We conclude that SHIV, regardless of pathogenicity, can cause injury to the small intestine and leads to CD4(+) T-cell depletion in infected animals during acute infection.
Collapse
|
24
|
Akiyama H, Ishimatsu M, Miura T, Hayami M, Ido E. Construction and infection of a new simian/human immunodeficiency chimeric virus (SHIV) containing the integrase gene of the human immunodeficiency virus type 1 genome and analysis of its adaptation to monkey cells. Microbes Infect 2008; 10:531-9. [PMID: 18403228 DOI: 10.1016/j.micinf.2008.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 01/29/2008] [Accepted: 02/04/2008] [Indexed: 11/17/2022]
Abstract
Expanding the HIV-1-derived regions in the SHIV genome may help to clarify the viral restriction factors determining the host range. In this study, we constructed a new SHIV having the reverse transcriptase and integrase-encoding regions of HIV-1 in addition to the 3' half genomic region of HIV-1. This SHIV, termed SHIVrti/3rn, could replicate in a monkey CD4+ T cell line, HSC-F, although its replication in monkey PBMCs was very weak. After SHIVrti/3rn was passaged in HSC-F cells for 26weeks, it gradually began to replicate in monkey PBMCs. This monkey-cell-adapted virus, termed SHIVrti/3rnP, could replicate in rhesus macaques. The whole genome of SHIVrti/3rnP was sequenced and was found to differ from SHIVrti/3rn at eleven positions. We constructed a series of mutants having some or all of these mutations and investigated their replication kinetics. The mutational analysis revealed that all of the mutations, but mainly the mutations in env, were responsible for the adaptation in HSC-F cells and were enough to replicate in rhesus PBMCs. Of all the SHIVs reported so far that can infect rhesus monkeys in vivo, SHIVrti/3rnP is the one that is genetically the closest to HIV-1.
Collapse
Affiliation(s)
- Hisashi Akiyama
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoin-Kawaracho, Kyoto 606-8507, Japan
| | | | | | | | | |
Collapse
|
25
|
Borsetti A, Baroncelli S, Maggiorella MT, Bellino S, Moretti S, Sernicola L, Belli R, Ridolfi B, Farcomeni S, Negri DRM, Cafaro A, Ensoli B, Titti F. Viral outcome of simian–human immunodeficiency virus SHIV-89.6P adapted to cynomolgus monkeys. Arch Virol 2007; 153:463-72. [DOI: 10.1007/s00705-007-0009-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 11/29/2007] [Indexed: 10/22/2022]
|
26
|
Kuwata T, Kodama M, Sato A, Suzuki H, Miyazaki Y, Miura T, Hayami M. Contribution of monocytes to viral replication in macaques during acute infection with simian immunodeficiency virus. AIDS Res Hum Retroviruses 2007; 23:372-80. [PMID: 17411370 DOI: 10.1089/aid.2006.0208] [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] [Indexed: 11/13/2022] Open
Abstract
Monocytes are known as an alternative target for HIV/SIV infection, but the contribution of monocytes to viral spread in a host is unclear. In this study, CD14 monocytes were monitored in 6 macaques until six weeks postinfection (wpi) with SIVmac239 to evaluate their contribution to viral load. The monocyte count in blood significantly increased with peak viremia at 2 wpi and the expression level of CD14 on monocytes significantly decreased at 1-2 wpi, though the number of CD4(+) T cells was stable in these macaques. The number of CD14 monocytes and the expression level of CD14 on monocytes at 2 wpi were also significantly related to the extent of viremia in plasma. An increased number of monocytes at 2 wpi was associated with a lower postacute viral load, suggesting that monocytes have a role in suppressing the virus. The lower expression level of CD14 in monocytes at 2 wpi was associated with a higher viral load and greater degree of infection of monocytes. This correlation suggests that monocytes with a low level of CD14 may be more susceptible to SIV and may enhance viral replication. The analysis of monocytes in persistently infected macaques revealed that the expression level of CD14 was also significantly low during persistent infection compared with naïve macaques, though the monocyte count was within the normal range. Monocytes may suppress viruses, perhaps by their immune function, during acute infection. However, infection of monocytes may increase the viral load and spread viruses in a host.
Collapse
Affiliation(s)
- Takeo Kuwata
- Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Shimizu Y, Inaba K, Kaneyasu K, Ibuki K, Himeno A, Okoba M, Goto Y, Hayami M, Miura T, Haga T. A genetically engineered live-attenuated simian-human immunodeficiency virus that co-expresses the RANTES gene improves the magnitude of cellular immunity in rhesus macaques. Virology 2006; 361:68-79. [PMID: 17157892 DOI: 10.1016/j.virol.2006.10.050] [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: 07/26/2006] [Revised: 08/22/2006] [Accepted: 10/28/2006] [Indexed: 11/18/2022]
Abstract
Regulated-on-activation-normal-T-cell-expressed-and-secreted (RANTES), a CC-chemokine, enhances antigen-specific T helper (Th) type-1 responses against HIV-1. To evaluate the adjuvant effects of RANTES against HIV vaccine candidate in SHIV-macaque models, we genetically engineered a live-attenuated SHIV to express the RANTES gene (SHIV-RANTES) and characterized the virus's properties in vivo. After the vaccination, the plasma viral loads were same in the SHIV-RANTES-inoculated monkeys and the parental nef-deleted SHIV (SHIV-NI)-inoculated monkeys. SHIV-RANTES provided some immunity in monkeys by remarkably increasing the antigen-specific CD4+ Th cell-proliferative response and by inducing an antigen-specific IFN-gamma ELISpot response. The magnitude of the immunity in SHIV-RANTES-immunized animals, however, failed to afford greater protection against a heterologous pathogenic SHIV (SHIV-C2/1) challenge compared to control SHIV-NI-immunized animals. SHIV-RANTES immunized monkeys, elicited robust cellular CD4+ Th responses and IFN-gamma ELISpot responses after SHIV-C2/1 challenge. These findings suggest that the chemokine RANTES can augment vaccine-elicited, HIV-specific CD4+ T cell responses.
Collapse
Affiliation(s)
- Yuya Shimizu
- Department of Veterinary Microbiology, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Watanabe S, Terashima K, Ohta S, Horibata S, Yajima M, Shiozawa Y, Dewan MZ, Yu Z, Ito M, Morio T, Shimizu N, Honda M, Yamamoto N. Hematopoietic stem cell–engrafted NOD/SCID/IL2Rγnull mice develop human lymphoid systems and induce long-lasting HIV-1 infection with specific humoral immune responses. Blood 2006; 109:212-8. [PMID: 16954502 DOI: 10.1182/blood-2006-04-017681] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractCritical to the development of an effective HIV/AIDS model is the production of an animal model that reproduces long-lasting active replication of HIV-1 followed by elicitation of virus-specific immune responses. In this study, we constructed humanized nonobese diabetic/severe combined immunodeficiency (NOD/SCID)/interleukin-2 receptor γ-chain knockout (IL2Rγnull) (hNOG) mice by transplanting human cord blood–derived hematopoietic stem cells that eventually developed into human B cells, T cells, and other monocytes/macrophages and 4 dendritic cells associated with the generation of lymphoid follicle–like structures in lymphoid tissues. Expressions of CXCR4 and CCR5 antigens were recognized on CD4+ cells in peripheral blood, the spleen, and bone marrow, while CCR5 was not detected on thymic CD4+ T cells. The hNOG mice showed marked, long-lasting viremia after infection with both CCR5- and CXCR4-tropic HIV-1 isolates for more than the 40 days examined, with R5 virus–infected animals showing high levels of HIV-DNA copies in the spleen and bone marrow, and X4 virus–infected animals showing high levels of HIV-DNA copies in the thymus and spleen. Furthermore, we detected both anti–HIV-1 Env gp120– and Gag p24–specific antibodies in animals showing a high rate of viral infection. Thus, the hNOG mice mirror human systemic HIV infection by developing specific antibodies, suggesting that they may have potential as an HIV/AIDS animal model for the study of HIV pathogenesis and immune responses.
Collapse
MESH Headings
- Animals
- Bone Marrow/pathology
- Bone Marrow/virology
- CD4-Positive T-Lymphocytes/virology
- Cell Lineage
- Cord Blood Stem Cell Transplantation
- DNA, Viral/analysis
- Disease Models, Animal
- Disease Susceptibility
- Female
- HIV Antibodies/biosynthesis
- HIV Antibodies/blood
- HIV Infections/immunology
- Humans
- Interleukin Receptor Common gamma Subunit/deficiency
- Interleukin Receptor Common gamma Subunit/genetics
- Lymphocyte Subsets/pathology
- Lymphoid Tissue/pathology
- Lymphoid Tissue/virology
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Receptors, CXCR4/analysis
- Receptors, CXCR4/genetics
- Receptors, CXCR5
- Receptors, Chemokine/analysis
- Receptors, Chemokine/genetics
- Spleen/pathology
- Spleen/virology
- Thymus Gland/pathology
- Thymus Gland/virology
- Transplantation, Heterologous
- Viremia/immunology
Collapse
Affiliation(s)
- Satoru Watanabe
- Department of Virology, Division of Medical Science, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Eda Y, Murakami T, Ami Y, Nakasone T, Takizawa M, Someya K, Kaizu M, Izumi Y, Yoshino N, Matsushita S, Higuchi H, Matsui H, Shinohara K, Takeuchi H, Koyanagi Y, Yamamoto N, Honda M. Anti-V3 humanized antibody KD-247 effectively suppresses ex vivo generation of human immunodeficiency virus type 1 and affords sterile protection of monkeys against a heterologous simian/human immunodeficiency virus infection. J Virol 2006; 80:5563-70. [PMID: 16699037 PMCID: PMC1472178 DOI: 10.1128/jvi.02095-05] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Accepted: 03/09/2006] [Indexed: 11/20/2022] Open
Abstract
In an accompanying report (Y. Eda, M. Takizawa, T. Murakami, H. Maeda, K. Kimachi, H. Yonemura, S. Koyanagi, K. Shiosaki, H. Higuchi, K. Makizumi, T. Nakashima, K. Osatomi, S. Tokiyoshi, S. Matsushita, N. Yamamoto, and M. Honda, J. Virol. 80:5552-5562, 2006), we discuss our production of a high-affinity humanized monoclonal antibody, KD-247, by sequential immunization with V3 peptides derived from human immunodeficiency virus type 1 (HIV-1) clade B primary isolates. Epitope mapping revealed that KD-247 recognized the Pro-Gly-Arg V3 tip sequence conserved in HIV-1 clade B isolates. In this study, we further demonstrate that in vitro, KD-247 efficiently neutralizes CXCR4- and CCR5-tropic primary HIV-1 clade B and clade B' with matching neutralization sequence motifs but does not neutralize sequence-mismatched clade B and clade E isolates. Monkeys were provided sterile protection against heterologous simian/human immunodeficiency virus challenge by the passive transfer of a single high dose (45 mg per kg of body weight) of KD-247 and afforded partial protection by lower antibody doses (30 and 15 mg per kg). Protective neutralization endpoint titers in plasma at the time of virus challenge were 1:160 in animals passively transferred with a high dose of the antibody. The antiviral efficacy of the antibody was further confirmed by its suppression of the ex vivo generation of primary HIV-1 quasispecies in peripheral blood mononuclear cell cultures from HIV-infected individuals. Therefore, KD-247 promises to be a valuable tool not only as a passive immunization antibody for the prevention of HIV infection but also as an immunotherapy for the suppression of HIV in phenotype-matched HIV-infected individuals.
Collapse
Affiliation(s)
- Yasuyuki Eda
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Eda Y, Takizawa M, Murakami T, Maeda H, Kimachi K, Yonemura H, Koyanagi S, Shiosaki K, Higuchi H, Makizumi K, Nakashima T, Osatomi K, Tokiyoshi S, Matsushita S, Yamamoto N, Honda M. Sequential immunization with V3 peptides from primary human immunodeficiency virus type 1 produces cross-neutralizing antibodies against primary isolates with a matching narrow-neutralization sequence motif. J Virol 2006; 80:5552-62. [PMID: 16699036 PMCID: PMC1472165 DOI: 10.1128/jvi.02094-05] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Accepted: 03/09/2006] [Indexed: 11/20/2022] Open
Abstract
An antibody response capable of neutralizing not only homologous but also heterologous forms of the CXCR4-tropic human immunodeficiency virus type 1 (HIV-1) MNp and CCR5-tropic primary isolate HIV-1 JR-CSF was achieved through sequential immunization with a combination of synthetic peptides representing HIV-1 Env V3 sequences from field and laboratory HIV-1 clade B isolates. In contrast, repeated immunization with a single V3 peptide generated antibodies that neutralized only type-specific laboratory-adapted homologous viruses. To determine whether the cross-neutralization response could be attributed to a cross-reactive antibody in the immunized animals, we isolated a monoclonal antibody, C25, which neutralized the heterologous primary viruses of HIV-1 clade B. Furthermore, we generated a humanized monoclonal antibody, KD-247, by transferring the genes of the complementary determining region of C25 into genes of the human V region of the antibody. KD-247 bound with high affinity to the "PGR" motif within the HIV-1 Env V3 tip region, and, among the established reference antibodies, it most effectively neutralized primary HIV-1 field isolates possessing the matching neutralization sequence motif, suggesting its promise for clinical applications involving passive immunizations. These results demonstrate that sequential immunization with B-cell epitope peptides may contribute to a humoral immune-based HIV vaccine strategy. Indeed, they help lay the groundwork for the development of HIV-1 vaccine strategies that use sequential immunization with biologically relevant peptides to overcome difficulties associated with otherwise poorly immunogenic epitopes.
Collapse
Affiliation(s)
- Yasuyuki Eda
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Miyake A, Ibuki K, Enose Y, Suzuki H, Horiuchi R, Motohara M, Saito N, Nakasone T, Honda M, Watanabe T, Miura T, Hayami M. Rapid dissemination of a pathogenic simian/human immunodeficiency virus to systemic organs and active replication in lymphoid tissues following intrarectal infection. J Gen Virol 2006; 87:1311-1320. [PMID: 16603534 DOI: 10.1099/vir.0.81307-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A better understanding of virological events during the early phase of human immunodeficiency virus 1 (HIV-1) infection is important for development of effective antiviral vaccines. In this study, by using quantitative PCR and an infectious plaque assay, virus distribution and replication were examined in various internal organs of rhesus macaques for almost 1 month after intrarectal inoculation of a pathogenic simian immunodeficiency virus/HIV chimeric virus (SHIV-C2/1-KS661c). At 3 days post-inoculation (p.i.), proviral DNA was detected in the rectum, thymus and axillary lymph node. In lymphoid tissues, infectious virus was first detected at 6 days p.i. and a high level of proviral DNA and infectious virus were both detected at 13 days p.i. By 27 days p.i., levels of infectious virus decreased dramatically, although proviral DNA load remained unaltered. In the intestinal tract, levels of infectious virus detected were much lower than in lymphoid tissues, whereas proviral DNA was detected at the same level as in lymphoid tissues throughout the infection. In the thymus and jejunum, CD4CD8 double-positive T cells were depleted earlier than CD4 single-positive cells. These results show that the virus spread quickly to systemic tissues after mucosal transmission. Thereafter, infectious virus was actively produced in the lymphoid tissues, but levels decreased significantly after the peak of viraemia. In contrast, in the intestinal tract, infectious virus was produced at low levels from the beginning of infection. Moreover, virus pathogenesis differed in CD4 single-positive and CD4CD8 double-positive T cells.
Collapse
Affiliation(s)
- Ariko Miyake
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Ibuki
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshimi Enose
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hajime Suzuki
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Reii Horiuchi
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Makiko Motohara
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoki Saito
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | | | - Mitsuo Honda
- National Institute of Infectious Disease, Tokyo 162-8640, Japan
| | - Toshiki Watanabe
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tomoyuki Miura
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanori Hayami
- Institute for Virus Research, Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
32
|
Horiuchi R, Akahata W, Kuwata T, Enose Y, Ido E, Suzuki H, Miyake A, Saito N, Ibuki K, Goto T, Miura T, Hayami M. DNA vaccination of macaques by a full-genome SHIV plasmid that has an IL-2 gene and produces non-infectious virus particles. Vaccine 2006; 24:3677-85. [PMID: 16085341 DOI: 10.1016/j.vaccine.2005.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 11/10/2004] [Accepted: 06/10/2005] [Indexed: 11/30/2022]
Abstract
We previously reported that a mutant full-sized plasmid DNA vaccine regime in macaques was effective against a homologous challenge [Akahata W, Ido E, Shimada T, Katsuyama K, Yamamoto H, Uesaka H, et al. DNA vaccination of macaques by a full genome HIV-1 plasmid which produces non-infectious virus particles. Virology 2000;275:116-24; Akahata W, Ido E, Akiyama H, Uesaka H, Enose Y, Horiuchi R, et al. DNA vaccination of macaques by a full genome SHIV-1 plasmid that produces non-infectious virus particles. J Gen Virol 2003;84:2237-44]. In this study, to evaluate the DNA vaccination regime against a heterologous challenge, a novel plasmid named pSHIV-ZF1*IL-2 was constructed. Four monkeys were intramuscularly and intradermally injected four times with the pSHIV-ZF1*IL-2. Vaccinated monkeys were intravenously challenged with a highly pathogenic, heterologous SHIV at 11 weeks post vaccination. All the vaccinated monkeys suppressed the challenge virus rapidly under the detectable level by 16 weeks post challenge. One vaccinated monkey was protected from a loss of CD4+ T cells. These results suggest pSHIV-ZF1*IL-2 alone seems partially effective even against a challenge with a heterologous, pathogenic virus.
Collapse
Affiliation(s)
- Reii Horiuchi
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Motohara M, Ibuki K, Miyake A, Fukazawa Y, Inaba K, Suzuki H, Masuda K, Minato N, Kawamoto H, Nakasone T, Honda M, Hayami M, Miura T. Impaired T-cell differentiation in the thymus at the early stages of acute pathogenic chimeric simian-human immunodeficiency virus (SHIV) infection in contrast to less pathogenic SHIV infection. Microbes Infect 2006; 8:1539-49. [PMID: 16702011 DOI: 10.1016/j.micinf.2006.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2005] [Revised: 01/12/2006] [Accepted: 01/16/2006] [Indexed: 10/24/2022]
Abstract
One of the mechanisms by which HIV infection induces the depletion of CD4+ T cells has been suggested to be impairment of T-cell development in the thymus, although there is no direct evidence that this occurs. To examine this possibility, we compared T-cell maturation in the intrathymic progenitors between macaques infected with an acute pathogenic chimeric simian-human immunodeficiency virus (SHIV), which causes profound and irreversible CD4+ T-cell depletion, and macaques infected with a less pathogenic SHIV, which causes only a transient CD4+ T-cell decline. Within 27 days post-inoculation (dpi), the two virus infections caused similar increases in plasma viral loads and similar decreases in CD4+ T-cell counts. However, in the thymus, the acute pathogenic SHIV resulted in increased thymic involution, atrophy and the depletion of immature T cells including CD4(+)CD8(+) double-positive (DP) cells, whereas the less pathogenic SHIV did not have these effects. Ex vivo differentiation of CD3(-)CD4(-)CD8(-) triple-negative (TN) intrathymic progenitors to DP cells was assessed by a monkey-mouse xenogenic fetal thymus organ culture system. Differentiation was impaired in the TN intrathymic progenitors of the acute pathogenic SHIV-infected monkeys, while differentiation was not impaired in the TN intrathymic progenitors of the less pathogenic SHIV-infected monkeys. These differences suggest that dysfunction of thymic maturation makes an important contribution to the irreversible depletion of circulating CD4+ T cells in vivo.
Collapse
Affiliation(s)
- Makiko Motohara
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, 53 Shogoinkawara-Machi, Sakyo-Ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Someya K, Ami Y, Nakasone T, Izumi Y, Matsuo K, Horibata S, Xin KQ, Yamamoto H, Okuda K, Yamamoto N, Honda M. Induction of Positive Cellular and Humoral Immune Responses by a Prime-Boost Vaccine Encoded with Simian Immunodeficiency Virusgag/pol. THE JOURNAL OF IMMUNOLOGY 2006; 176:1784-95. [PMID: 16424209 DOI: 10.4049/jimmunol.176.3.1784] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is believed likely that immune responses are responsible for controlling viral load and infection. In this study, when macaques were primed with plasmid DNA encoding SIV gag and pol genes (SIVgag/pol DNA) and then boosted with replication-deficient vaccinia virus DIs recombinant expressing the same genes (rDIsSIVgag/pol), this prime-boost regimen generated higher levels of Gag-specific CD4+ and CD8+ T cell responses than did either SIVgag/pol DNA or rDIsSIVgag/pol alone. When the macaques were i.v. challenged with pathogenic simian/HIV, the prime-boost group maintained high CD4+ T cell counts and reduced plasma viral loads up to 30 wk after viral challenge, whereas the rDIsSIVgag/pol group showed only a partial attenuation of the viral infection, and the group immunized with SIVgag/pol DNA alone showed none at all. The protection levels were better correlated with the levels of virus-specific T cell responses than the levels of neutralization Ab responses. These results demonstrate that a vaccine regimen that primes with DNA and then boosts with a replication-defective vaccinia virus DIs generates anti-SIV immunity, suggesting that it will be a promising vaccine regimen for HIV-1 vaccine development.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/blood
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Female
- Flow Cytometry
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Gene Products, pol/genetics
- Gene Products, pol/immunology
- Genetic Vectors
- Immunity, Cellular
- Immunization, Secondary
- Interferon-gamma/metabolism
- Kinetics
- Macaca fascicularis
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/prevention & control
- Simian Immunodeficiency Virus/genetics
- Simian Immunodeficiency Virus/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccinia virus
Collapse
Affiliation(s)
- Kenji Someya
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kawahara M, Matsuo K, Honda M. Intradermal and oral immunization with recombinant Mycobacterium bovis BCG expressing the simian immunodeficiency virus Gag protein induces long-lasting, antigen-specific immune responses in guinea pigs. Clin Immunol 2006; 119:67-78. [PMID: 16386958 DOI: 10.1016/j.clim.2005.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 10/14/2005] [Accepted: 11/09/2005] [Indexed: 11/17/2022]
Abstract
To develop a new recombinant BCG (rBCG) vaccine, we constructed rBCG that expresses the full-length Gag protein of simian immunodeficiency virus (rBCG-SIVGag) at a level of 0.5 ng/mg after 3 weeks of bacterial cell culture. Intradermal (i.d.) inoculation of guinea pigs with 0.1 mg of rBCG-SIVGag resulted in the induction of delayed-type hypersensitivity (DTH) responses to both purified protein derivative (PPD) of tuberculin and SIV Gag p27 protein; responses that were maintained for the duration of the 50-week study. In contrast, guinea pigs orally vaccinated with 160 mg of the same antigen exhibited a long-lasting DTH response to the SIV Gag p27 protein, but mounted no response to PPD. Proliferative responses to SIV Gag p27 and PPD antigens were detected in both i.d. and orally immunized animals; however, the levels of PPD-specific responses were significantly higher in guinea pigs immunized by the i.d. than the oral route. A significant increase in the level of PPD- and SIV Gag p27-specific IFNgamma mRNA expression was also detected in both immunization groups receiving rBCG-SIVGag. In addition, both i.d. and oral immunization with rBCG-SIVGag induced PPD- and SIV Gag p27-specific serum IgG responses. Insertion of the SIV gag gene into BCG did not appear to change the ability of rBCG-immunized animals to elicit PPD-specific immune responses. These results indicate that rBCG-SIVGag has the ability to effectively induce long-lasting, cell-mediated and humoral immunity against both viral and bacterial antigens in guinea pigs, suggesting that rBCG-Gag has the potential to elicit immunities specific not only for tuberculosis but also for HIV at human doses.
Collapse
Affiliation(s)
- Mamoru Kawahara
- National Institute of Infectious Diseases, 1-23-1 Toyama, Tokyo 162-8640, Japan.
| | | | | |
Collapse
|
36
|
Miyake A, Ibuki K, Suzuki H, Horiuchi R, Saito N, Motohara M, Hayami M, Miura T. Early virological events in various tissues of newborn monkeys after intrarectal infection with pathogenic simian human immunodeficiency virus. J Med Primatol 2005; 34:294-302. [PMID: 16128924 DOI: 10.1111/j.1600-0684.2005.00127.x] [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: 11/28/2022]
Abstract
Children infected with human immunodeficiency virus type 1 often have higher viral loads and progress to acquired immunodeficiency syndrome more rapidly than adults. In our previous study of simian-human immunodeficiency virus (SHIV)-infected adult monkeys, immature CD4CD8 double-positive T cells in the thymus and jejunum decreased faster than mature CD4 single-positive T cells. Here, we examined the effect of virus replication on immature T cells from the same SHIV-inoculated newborn monkeys having more immature T cells than adults. The infectious viruses were more abundantly detected in the thymus than in other tissues at both 13 and 26 days post-infection (dpi). However, mature CD4(+) T cells in the thymus declined after 13 dpi and immature CD3(-) CD4 single-positive T cells remained at 26 dpi. These results suggested that many immature CD4(+) T cells in the thymus of newborns support the production of infectious viruses even after the depletion of mature CD4(+) T cells.
Collapse
Affiliation(s)
- Ariko Miyake
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ami Y, Izumi Y, Matsuo K, Someya K, Kanekiyo M, Horibata S, Yoshino N, Sakai K, Shinohara K, Matsumoto S, Yamada T, Yamazaki S, Yamamoto N, Honda M. Priming-boosting vaccination with recombinant Mycobacterium bovis bacillus Calmette-Guérin and a nonreplicating vaccinia virus recombinant leads to long-lasting and effective immunity. J Virol 2005; 79:12871-9. [PMID: 16188989 PMCID: PMC1235843 DOI: 10.1128/jvi.79.20.12871-12879.2005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Virus-specific T-cell responses can limit immunodeficiency virus type 1 (HIV-1) transmission and prevent disease progression and so could serve as the basis for an affordable, safe, and effective vaccine in humans. To assess their potential for a vaccine, we used Mycobacterium bovis bacillus Calmette-Guérin (BCG)-Tokyo and a replication-deficient vaccinia virus strain (DIs) as vectors to express full-length gag from simian immunodeficiency viruses (SIVs) (rBCG-SIVgag and rDIsSIVgag). Cynomolgus macaques were vaccinated with either rBCG-SIVgag dermally as a single modality or in combination with rDIsSIVgag intravenously. When cynomologus macaques were primed with rBCG-SIVgag and then boosted with rDIsSIVgag, high levels of gamma interferon (IFN-gamma) spot-forming cells specific for SIV Gag were induced. This combination regimen elicited effective protective immunity against mucosal challenge with pathogenic simian-human immunodeficiency virus for the 1 year the macaques were under observation. Antigen-specific intracellular IFN-gamma activity was similarly induced in each of the macaques with the priming-boosting regimen. Other groups receiving the opposite combination or the single-modality vaccines were not effectively protected. These results suggest that a recombinant M. bovis BCG-based vector may have potential as an HIV/AIDS vaccine when administered in combination with a replication-deficient vaccinia virus DIs vector in a priming-boosting strategy.
Collapse
Affiliation(s)
- Yasushi Ami
- Division of Experimental Animal Research, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Suzuki H, Motohara M, Miyake A, Ibuki K, Fukazawa Y, Inaba K, Masuda K, Minato N, Kawamoto H, Hayami M, Miura T. Intrathymic effect of acute pathogenic SHIV infection on T-lineage cells in newborn macaques. Microbiol Immunol 2005; 49:667-79. [PMID: 16034211 DOI: 10.1111/j.1348-0421.2005.tb03646.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We intrarectally infected newborn macaques with a pathogenic simian/human immunodeficiency virus (SHIV) that induced rapid and profound CD4 (+) T cell depletion, and examined the early effects of this SHIV on the thymus. After intrarectal infection, viral loads were much higher in the thymus than in other lymphoid tissues in newborns. In contrast, no clear difference was seen in the viral loads of different tissues in adults. Histological and immunohistochemical observations showed severe thymic involution. Depletion of CD4 (+) thymocytes began in the medulla at 2 weeks post infection and spread over the whole thymus. After in vivo infection, the CD2 (+) subpopulation, which represents a relatively later stage of T cell progenitors, was selectively reduced and development of thymocytes from CD3 (-) CD4 (-) CD8 (-) cells to CD4 (+) CD8 (+) cells was impaired. These results suggest that profound and irreversible loss of CD4 (+) cells that are observed in the peripheral blood of SHIV-infected monkeys are due to destruction of the thymus and impaired thymopoiesis as a result of SHIV infection in the thymus.
Collapse
Affiliation(s)
- Hajime Suzuki
- Laboratory of Primate Model, Institute for Virus Research, Kyoto University, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Shimizu Y, Miyazaki Y, Ibuki K, Suzuki H, Kaneyasu K, Goto Y, Hayami M, Miura T, Haga T. Induction of immune response in macaque monkeys infected with simian-human immunodeficiency virus having the TNF-alpha gene at an early stage of infection. Virology 2005; 343:151-61. [PMID: 16169034 DOI: 10.1016/j.virol.2005.08.025] [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: 04/23/2005] [Revised: 06/07/2005] [Accepted: 08/18/2005] [Indexed: 12/21/2022]
Abstract
TNF-alpha has been implicated in the pathogenesis of, and the immune response against, HIV-1 infection. To clarify the roles of TNF-alpha against HIV-1-related virus infection in an SHIV-macaque model, we genetically engineered an SHIV to express the TNF-alpha gene (SHIV-TNF) and characterized the virus's properties in vivo. After the acute viremic stage, the plasma viral loads declined earlier in the SHIV-TNF-inoculated monkeys than in the parental SHIV (SHIV-NI)-inoculated monkeys. SHIV-TNF induced cell death in the lymph nodes without depletion of circulating CD4(+) T cells. SHIV-TNF provided some immunity in monkeys by increasing the production of the chemokine RANTES and by inducing an antigen-specific proliferation of lymphocytes. The monkeys immunized with SHIV-TNF were partly protected against a pathogenic SHIV (SHIV-C2/1) challenge. These findings suggest that TNF-alpha contributes to the induction of an effective immune response against HIV-1 rather than to the progression of disease at the early stage of infection.
Collapse
Affiliation(s)
- Yuya Shimizu
- Department of Veterinary Microbiology, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Someya K, Cecilia D, Ami Y, Nakasone T, Matsuo K, Burda S, Yamamoto H, Yoshino N, Kaizu M, Ando S, Okuda K, Zolla-Pazner S, Yamazaki S, Yamamoto N, Honda M. Vaccination of rhesus macaques with recombinant Mycobacterium bovis bacillus Calmette-Guérin Env V3 elicits neutralizing antibody-mediated protection against simian-human immunodeficiency virus with a homologous but not a heterologous V3 motif. J Virol 2005; 79:1452-62. [PMID: 15650171 PMCID: PMC544111 DOI: 10.1128/jvi.79.3.1452-1462.2005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although the correlates of vaccine-induced protection against human immunodeficiency virus type 1 (HIV-1) are not fully known, it is presumed that neutralizing antibodies (NAb) play a role in controlling virus infection. In this study, we examined immune responses elicited in rhesus macaques following vaccination with recombinant Mycobacterium bovis bacillus Calmette-Guérin expressing an HIV-1 Env V3 antigen (rBCG Env V3). We also determined the effect of vaccination on protection against challenge with either a simian-human immunodeficiency virus (SHIV-MN) or a highly pathogenic SHIV strain (SHIV-89.6PD). Immunization with rBCG Env V3 elicited significant levels of NAb for the 24 weeks tested that were predominantly HIV-1 type specific. Sera from the immunized macaques neutralized primary HIV-1 isolates in vitro, including HIV-1BZ167/X4, HIV-1SF2/X4, HIV-1CI2/X4, and, to a lesser extent, HIV-1MNp/X4, all of which contain a V3 sequence homologous to that of rBCG Env V3. In contrast, neutralization was not observed against HIV-1SF33/X4, which has a heterologous V3 sequence, nor was it found against primary HIV-1 R5 isolates from either clade A or B. Furthermore, the viral load in the vaccinated macaques was significantly reduced following low-dose challenge with SHIV-MN, and early plasma viremia was markedly decreased after high-dose SHIV-MN challenge. In contrast, replication of pathogenic SHIV-89.6PD was not affected by vaccination in any of the macaques. Thus, we have shown that immunization with an rBCG Env V3 vaccine elicits a strong, type-specific V3 NAb response in rhesus macaques. While this response was not sufficient to provide protection against a pathogenic SHIV challenge, it was able to significantly reduce the viral load in macaques following challenge with a nonpathogenic SHIV. These observations suggest that rBCG vectors have the potential to deliver an appropriate virus immunogen for desirable immune elicitations.
Collapse
Affiliation(s)
- Kenji Someya
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yamakami K, Honda M, Takei M, Ami Y, Kitamura N, Nishinarita S, Sawada S, Horie T. Early bone marrow hematopoietic defect in simian/human immunodeficiency virus C2/1-infected macaques and relevance to advance of disease. J Virol 2004; 78:10906-10. [PMID: 15452210 PMCID: PMC521857 DOI: 10.1128/jvi.78.20.10906-10910.2004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To clarify hematological abnormalities following infection with human immunodeficiency virus (HIV), we examined the hematopoietic capability of bone marrow by using cynomolgus monkeys infected with pathogenic simian/human immunodeficiency virus (SHIV) strain C2/1, an animal model of HIV infection. The relationship between the progress of the infection and the CD4/CD8 ratio of T lymphocytes or the amount of SHIV C2/1 viral load in the peripheral blood was also investigated. A colony assay was performed to assess the hematopoietic capability of bone marrow stem cells during the early and advanced phases of the infection. Colonies of granulocytes-macrophages (GM) were examined by PCR for the presence of the SIVmac239 gag region to reveal direct viral infection. There was a remarkable decrease in the CFU-GM growth on days 1 and 3 postinoculation, followed by recovery on day 56. During the more advanced stage, the CFU-GM growth decreased again. There was minimal evidence of direct viral infection of pooled cultured CFU-GM despite the continuously low CD4/CD8 ratios. These results indicate that the decrease in colony formation by bone marrow stem cells is reversible and fluctuates with the advance of the disease. This decrease was not due to direct viral infection of CFU-GM. Our data may support the concept that, in the early phase, production of inhibitory factors or deficiency of a stimulatory cytokine is responsible for some of the bone marrow defects described in the SHIV C2/1 model.
Collapse
Affiliation(s)
- Kenji Yamakami
- Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Someya K, Xin KQ, Matsuo K, Okuda K, Yamamoto N, Honda M. A consecutive priming-boosting vaccination of mice with simian immunodeficiency virus (SIV) gag/pol DNA and recombinant vaccinia virus strain DIs elicits effective anti-SIV immunity. J Virol 2004; 78:9842-53. [PMID: 15331719 PMCID: PMC515009 DOI: 10.1128/jvi.78.18.9842-9853.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To evaluate immunity induced by a novel DNA prime-boost regimen, we constructed a DNA plasmid encoding the gag and pol genes from simian immunodeficiency virus (SIV) (SIVgag/pol DNA), in addition to a replication-deficient vaccinia virus strain DIs recombinant expressing SIV gag and pol genes (rDIsSIVgag/pol). In mice, priming with SIVgag/pol DNA, followed by rDIsSIVgag/pol induced an SIV-specific lymphoproliferative response that was mediated by a CD4+-T-lymphocyte subset. Immunization with either vaccine alone was insufficient to induce high levels of proliferation or Th1 responses in the animals. The prime-boost regimen also induced SIV Gag-specific cellular responses based on gamma interferon secretion, as well as cytotoxic-T-lymphocyte responses. Thus, the regimen of DNA priming and recombinant DIs boosting induced Th1-type cell-mediated immunity, which was associated with resistance to viral challenge with wild-type vaccinia virus expressing SIVgag/pol, suggesting that this new regimen may hold promise as a safe and effective vaccine against human immunodeficiency virus type 1.
Collapse
Affiliation(s)
- Kenji Someya
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Izumi Y, Ami Y, Matsuo K, Someya K, Sata T, Yamamoto N, Honda M. Intravenous inoculation of replication-deficient recombinant vaccinia virus DIs expressing simian immunodeficiency virus gag controls highly pathogenic simian-human immunodeficiency virus in monkeys. J Virol 2004; 77:13248-56. [PMID: 14645581 PMCID: PMC296093 DOI: 10.1128/jvi.77.24.13248-13256.2003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To be effective, a vaccine against human immunodeficiency virus type 1 (HIV-1) must induce virus-specific T-cell responses and it must be safe for use in humans. To address these issues, we developed a recombinant vaccinia virus DIs vaccine (rDIsSIVGag), which is nonreplicative in mammalian cells and expresses the full-length gag gene of simian immunodeficiency virus (SIV). Intravenous inoculation of 10(6) PFU of rDIsSIVGag in cynomologus macaques induced significant levels of gamma interferon (IFN-gamma) spot-forming cells (SFC) specific for SIV Gag. Antigen-specific lymphocyte proliferative responses were also induced and were temporally associated with the peak of IFN-gamma SFC activity in each macaque. In contrast, macaques immunized with a vector control (rDIsLacZ) showed no significant induction of antigen-specific immune responses. After challenge with a highly pathogenic simian-human immunodeficiency virus (SHIV), CD4(+) T lymphocytes were maintained in the peripheral blood and lymphoid tissues of the immunized macaques. The viral set point in plasma was also reduced in these animals, which may be related to the enhancement of virus-specific intracellular IFN-gamma(+) CD8(+) cell numbers and increased antibody titers after SHIV challenge. These results demonstrate that recombinant DIs has potential for use as an HIV/AIDS vaccine.
Collapse
Affiliation(s)
- Yasuyuki Izumi
- AIDS Research Center. Division of Experimental Animal Research. Department of Pathology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640. Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Miyake A, Akagi T, Enose Y, Ueno M, Kawamura M, Horiuchi R, Hiraishi K, Adachi M, Serizawa T, Narayan O, Akashi M, Baba M, Hayami M. Induction of HIV-specific antibody response and protection against vaginal SHIV transmission by intranasal immunization with inactivated SHIV-capturing nanospheres in macaques. J Med Virol 2004; 73:368-77. [PMID: 15170630 DOI: 10.1002/jmv.20100] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We have previously reported that concanavalin A-immobilized polystyrene nanospheres (Con A-NS) could efficiently capture HIV-1 particles and that intranasal immunization with inactivated HIV-1-capturing nanospheres (HIV-NS) induced vaginal anti-HIV-1 IgA antibody response in mice. In this study, to evaluate the protective effect of immunization, each three macaques was intranasally immunized with Con A-NS or inactivated simian/human immunodeficiency virus KU-2-capturing nanospheres (SHIV-NS) and then intravaginally challenged with a pathogenic virus, SHIV KU-2. After a series of six immunizations, vaginal anti-HIV-1 gp120 IgA and IgG antibodies were detected in all SHIV-NS-immunized macaques. After intravaginal challenge, one of the three macaques in each of the Con A-NS- and SHIV-NS-immunized groups was infected. Plasma viral RNA load of infected macaque in SHIV-NS-immunized macaques was substantially less than that in unimmunized control macaque and reached below the detectable level. However, it could not be determined whether intranasal immunization with SHIV-NS is effective in giving complete protection against intravaginal challenge. To explore the effect of the SHIV-NS vaccine, the remaining non-infected macaques were rechallenged intravenously with SHIV KU-2. After intravenous challenge, all macaques became infected. However, SHIV-NS-immunized macaques had lower viral RNA loads and higher CD4(+) T cell counts than unimmunized control macaques. Plasma anti-HIV-1 gp120 IgA and IgG antibodies were induced more rapidly in the SHIV-NS-immunized macaques than in the controls. The rapid antibody responses having neutralizing activity might contribute to the clearance of the challenge virus. Thus, SHIV-NS-immunized macaques exhibited partial protection to vaginal and systemic challenges with SHIV KU-2.
Collapse
Affiliation(s)
- Ariko Miyake
- Laboratory of Primate Model, Experimental Research Center for Infectious Disease, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kaizu M, Ami Y, Nakasone T, Sasaki Y, Izumi Y, Sato H, Takahashi E, Sakai K, Shinohara K, Nakanishi K, Honda M. Higher levels of IL-18 circulate during primary infection of monkeys with a pathogenic SHIV than with a nonpathogenic SHIV. Virology 2003; 313:8-12. [PMID: 12951016 DOI: 10.1016/s0042-6822(03)00434-3] [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] [Indexed: 11/17/2022]
Abstract
We have monitored kinetics of peripheral blood Interleukin (IL)-18 level, viral RNA load, and CD4(+) T cell counts in cynomolgus and rhesus macaques following infections of various simian/human immunodeficiency viruses (SHIVs) causing differential pathogenicity. Infections of cynomolgus and rhesus macaques with pathogenic SHIVs-C2/1 and -89.6PD, respectively, induced high levels of plasma IL-18 (0.1-1 ng/ml) and enhanced apoptosis of peripheral blood T cells during primary viremia, along with a rapid decline of CD4(+) T cells and a high level of set point viral load after primary viremia (six of six cases). In contrast, infections of cynomolgus macaques with nonpathogenic SHIVs-TH09V3 and -MD14 did not cause such IL-18 elevation, showing no decline of CD4(+) T cells and no or low viral set point level following primary viremia (three of three cases). Thus, the elevation of circulating IL-18 level during primary viral infection can be a good indicator of an active pathogenic viral infection. However, the role of increased IL-18 remains to be elucidated and needs further investigation.
Collapse
Affiliation(s)
- Masahiko Kaizu
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ishii K, Ueda Y, Matsuo K, Matsuura Y, Kitamura T, Kato K, Izumi Y, Someya K, Ohsu T, Honda M, Miyamura T. Structural analysis of vaccinia virus DIs strain: application as a new replication-deficient viral vector. Virology 2002; 302:433-44. [PMID: 12441087 DOI: 10.1006/viro.2002.1622] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
DIs is a restrictive host range mutant of vaccinia virus strain DIE that grows well only in chick embryo fibroblast cells but is unable to grow in most mammalian cells. In this study, we identified one major deletion (15.4 kbp) which results in the loss of 19 putative open reading frames in the left end of the genome. We then established a system to express foreign genes by inserting them into the deleted region of DIs. We constructed rDIs to express the bacteriophage T7 polymerase (T7pol) gene and showed the expression in various mammalian cell lines by reporter luciferase gene expression under the T7 promoter. We also expressed the full-length human immunodeficiency virus (HIV)-1 NL432 gag gene. The expressed gag gene product induced high levels of cytotoxic T lymphocytes in immunized mice. These data suggest that DIs is useful as an efficient, transient replication-deficient viral vector.
Collapse
Affiliation(s)
- Koji Ishii
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kojima Y, Xin KQ, Ooki T, Hamajima K, Oikawa T, Shinoda K, Ozaki T, Hoshino Y, Jounai N, Nakazawa M, Klinman D, Okuda K. Adjuvant effect of multi-CpG motifs on an HIV-1 DNA vaccine. Vaccine 2002; 20:2857-65. [PMID: 12126895 DOI: 10.1016/s0264-410x(02)00238-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs trigger an immune response characterized by the activation of B cells, NK cells and monocytes/macrophages. Based on evidence that the immunogenicity of DNA vaccines can be augmented by the addition of CpG motifs, 5-20 additional CpG motifs were cloned into a pUC-derived plasmid. Treating bone-marrow derived dendritic cells (BM-DCs) with CpG-enriched plasmids in vitro boosted their expressions of MHC class II molecules, the CD40 and CD86 activation markers. Co-administering the CpG-enriched plasmids with a DNA vaccine encoding the envelope glycoprotein of HIV to BALB/c mice significantly increased HIV-specific cell mediated and humoral immunity. A significant boost was observed when the CpG plasmid was administered either 2 or 4 days after DNA vaccination. Plasmids containing 20 CpG copies were the most effective immune enhancers both in vitro and in vivo. These results suggest that plasmids containing multiple CpG motifs may improve the immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- Yoshitsugu Kojima
- Department of Bacteriology, University School of Medicine, Yokohama City, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Plemper RK, Hammond AL, Gerlier D, Fielding AK, Cattaneo R. Strength of envelope protein interaction modulates cytopathicity of measles virus. J Virol 2002; 76:5051-61. [PMID: 11967321 PMCID: PMC136138 DOI: 10.1128/jvi.76.10.5051-5061.2002] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To understand the molecular determinants of measles virus (MV) cytopathicity, we have characterized mutant viruses exhibiting a more-extensive cell-to-cell fusion while maintaining efficient replication to high titers. A virus which is modified by the addition of an 8-amino-acid Flag epitope tag at the cytoplasmic tail of its H (for MV hemagglutinin) envelope glycoprotein replicates efficiently, has an increased cytopathicity, possesses a greater infectivity per particle, and has an altered protein composition compared with that of unmodified MV. The mutant phenotype is not specifically linked to the epitope sequence, since an alternatively added HA (for influenza virus-derived hemagglutinin) epitope tag caused similar effects. We demonstrate that both epitope tags weaken the interaction between the H and fusion (F) glycoproteins in virus-infected cells. This reduction in strength of H/F interaction is independent of the presence of the viral matrix (M) protein. Viruses with this less stable complex are more sensitive to neutralization by a soluble octameric form of the CD46 receptor, consistent with their increased fusogenicity. Similar analyses of glycoproteins derived from MV strains with reduced cytopathicities confirm that the strength of H and F glycoprotein interaction is a modulator of viral fusogenicity.
Collapse
Affiliation(s)
- Richard K Plemper
- Molecular Medicine Program, Mayo Foundation, Rochester, Minnesota 55905, USA.
| | | | | | | | | |
Collapse
|
49
|
Johnston JB, Power C. Feline immunodeficiency virus xenoinfection: the role of chemokine receptors and envelope diversity. J Virol 2002; 76:3626-36. [PMID: 11907202 PMCID: PMC136059 DOI: 10.1128/jvi.76.8.3626-3636.2002] [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: 11/20/2022] Open
Abstract
The use of chemokine receptors as cell recognition signals is a property common to several lentiviruses, including feline, human, and simian immunodeficiency viruses. Previously, two feline immunodeficiency virus (FIV) isolates, V1CSF and Petaluma, were shown to use chemokine receptors in a strain-dependent manner to infect human peripheral blood mononuclear cells (PBMC) (J. Johnston and C. Power, J. Virol. 73:2491-2498, 1999). Since the sequences of these viruses differed primarily in regions of the FIV envelope gene implicated in receptor use and cell tropism, envelope chimeras of V1CSF and Petaluma were constructed to investigate the role of envelope diversity in the profiles of chemokine receptors used by FIV to infect primate cells. By use of a receptor-blocking assay, all viruses were found to infect human and macaque PBMC through a mechanism involving the CXCR4 receptor. However, infection by viruses encoding the V3-to-V5 region of the V1CSF surface unit was also inhibited by blockade of the CCR3 or CCR5 receptor. Similar results were obtained with GHOST cells, human osteosarcoma cells expressing specific combinations of chemokine receptors. CXCR4 was required for infection by all FIV strains, but viruses expressing the V3-to-V5 region of V1CSF required the concurrent presence of either CCR3 or CCR5. In contrast, CXCR4 alone was sufficient to allow infection of GHOST cells by FIV strains possessing the V3-to-V5 region of Petaluma. To assess the role of primate chemokine receptors in productive infection, Crandell feline kidney (CrFK) cells that expressed human CXCR4, CCR3, or CCR5 in addition to feline CXCR4 were generated. Sustained infection by viruses encoding the V3-to-V5 region of V1CSF was detected in CrFK cells expressing human CCR3 or CCR5 but not in cells expressing CXCR4 alone, while all CrFK cell lines were permissive to viruses encoding the V3-to-V5 region of Petaluma. These results indicate that FIV uses chemokine receptors to infect both human and nonhuman primate cells and that the profiles of these receptors are dependent on envelope sequence, and they provide insights into the mechanism by which xenoinfections may occur.
Collapse
Affiliation(s)
- J B Johnston
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
50
|
Chujoh Y, Matsuo K, Yoshizaki H, Nakasatomi T, Someya K, Okamoto Y, Naganawa S, Haga S, Yoshikura H, Yamazaki A, Yamazaki S, Honda M. Cross-clade neutralizing antibody production against human immunodeficiency virus type 1 clade E and B' strains by recombinant Mycobacterium bovis BCG-based candidate vaccine. Vaccine 2001; 20:797-804. [PMID: 11738743 DOI: 10.1016/s0264-410x(01)00398-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The recombinant Mycobacterium bovis BCG (rBCG) vector-based vaccine secreting the V3 principal neutralizing epitope of human immunodeficiency virus type 1 (HIV-1) Japanese strain was reported to induce both humoral and cellular immune responses effectively [Proc. Natl. Acad. Sci. USA. 92 (1995) 10693]. The antigen-secreting rBCG system was applied to the V3 epitope of clade E HIV-1 in this study. The V3 sequence of 19 amino acids (aa) and 15aa fused with mycobacterial alpha-antigen was not secreted while 12aa and 11aa sequences were successfully secreted from BCG cells. Serum IgG from guinea pig which was immunized with 12aa epitope-secreting recombinant BCG neutralized the WHO reference strain as well as primary field isolates of clade E virus. The serum IgG could also neutralize Thai B (clade B') strains which possessed a conserved GPGQ motif in their V3 sequences. These data suggest that the rBCG construct secreting the 12aa epitope is implicated in the development of a prophylactic vaccine in Thailand in which both clade E and B' viruses are prevalent.
Collapse
Affiliation(s)
- Y Chujoh
- Central Research Laboratories, Ajinomoto Co., Inc., Suzuki-cho 1-1, Kawasaki-ku, Kawasaki 210-0801, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|