51
|
Lack of protection against feline immunodeficiency virus infection among domestic cats in New Zealand vaccinated with the Fel-O-Vax® FIV vaccine. Vet Microbiol 2020; 250:108865. [PMID: 33045631 DOI: 10.1016/j.vetmic.2020.108865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/17/2020] [Indexed: 11/20/2022]
Abstract
Infections with feline immunodeficiency virus (FIV) are common in New Zealand, although the impact of those infections on the health status of the cats remains unclear. Although many cats are vaccinated yearly with a commercial FIV vaccine containing FIV subtypes A and D, the effectiveness of this vaccine in protection against infection with field FIVs is unclear, as a high proportion of New Zealand viruses belong to subtype C. The objective of the study was to compare the frequency of FIV infection among adult FIV-vaccinated and FIV-unvaccinated domestic cats with access to outdoors. Buccal swabs were collected by the participating veterinarians and tested for the presence of FIV provirus by quantitative PCR. Overall, 26/185 (14.0 %) samples were positive for FIV, including 7/82 (8.5 %) samples from FIV-unvaccinated and 19/103 (18.4 %) from FIV-vaccinated cats. There was no protective effect of vaccination on FIV infection among sampled cats (p = 0.05). Partial sequences of the FIV envelope gene from five New Zealand viruses were analysed by the maximum likelihood method. All clustered with other New Zealand FIV sequences from subtypes A (n = 2), C (n = 2) or putative recombinant viruses (n = 1). While the FIV vaccination did not prevent FIV infection among sampled cats, it may have had an impact on transmissibility of the virus or on disease progression. As neither was addressed in the current study, further research is needed to fully assess the potential benefits of FIV vaccination. Considering the frequency of FIV infection in FIV-vaccinated cats, FIV infection status should be monitored not only before the first vaccination, but before each yearly booster.
Collapse
|
52
|
Challenges for the Newborn Immune Response to Respiratory Virus Infection and Vaccination. Vaccines (Basel) 2020; 8:vaccines8040558. [PMID: 32987691 PMCID: PMC7712002 DOI: 10.3390/vaccines8040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
The initial months of life reflect an extremely challenging time for newborns as a naïve immune system is bombarded with a large array of pathogens, commensals, and other foreign entities. In many instances, the immune response of young infants is dampened or altered, resulting in increased susceptibility and disease following infection. This is the result of both qualitative and quantitative changes in the response of multiple cell types across the immune system. Here we provide a review of the challenges associated with the newborn response to respiratory viral pathogens as well as the hurdles and advances for vaccine-mediated protection.
Collapse
|
53
|
Zang J, Gu C, Zhou B, Zhang C, Yang Y, Xu S, Bai L, Zhang R, Deng Q, Yuan Z, Tang H, Qu D, Lavillette D, Xie Y, Huang Z. Immunization with the receptor-binding domain of SARS-CoV-2 elicits antibodies cross-neutralizing SARS-CoV-2 and SARS-CoV without antibody-dependent enhancement. Cell Discov 2020; 6:61. [PMID: 32901211 PMCID: PMC7471522 DOI: 10.1038/s41421-020-00199-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 01/15/2023] Open
Affiliation(s)
- Jinkai Zang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenjian Gu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Bingjie Zhou
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Zhang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong Yang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiqi Xu
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lulu Bai
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Rong Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Qiang Deng
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Hong Tang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Di Qu
- BSL-3 Laboratory of Fudan University, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Zhong Huang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
54
|
Vangelista L, Secchi M. Prepare for the Future: Dissecting the Spike to Seek Broadly Neutralizing Antibodies and Universal Vaccine for Pandemic Coronaviruses. Front Mol Biosci 2020; 7:226. [PMID: 33033717 PMCID: PMC7490329 DOI: 10.3389/fmolb.2020.00226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 01/07/2023] Open
Abstract
Learning from the lengthy fight against HIV-1, influenza, and Ebola virus infection, broadly neutralizing antibodies (bnAbs), directed at conserved regions of surface proteins crucial to virus entry (Env, hemagglutinin, and GP, respectively), are an essential resource for passive as well as active immunization. Rare in their emergence and antigen recognition mode, bnAbs are active toward a large set of different viral strains. Isolation, characterization and production of bnAbs lead to their possible use in passive immunotherapy and form the basis for an educated effort in the development of vaccines for universal coverage. SARS-CoV-2-specific antibodies targeting the spike receptor binding domain (RBD) may lead to antibody dependent enhancement (ADE) of infection, possibly hampering the field of vaccine development. This perspective points to the identification of conserved regions in the spike of SARS-CoV-2, SARS-CoV, and MERS-CoV through investigation, dissection and recombinant production of isolated moieties. These spike moieties should be capable of independent folding and allow the detection as well as the elicitation of bnAbs, thus setting the basis for an effective passive immunotherapy and the development of a universal vaccine against human epidemic coronaviruses (HCoVs). SARS, MERS and, most of all, COVID-19 demonstrate that humanity is the target of HCoV, preparedness for future hits is thus no longer an option.
Collapse
Affiliation(s)
- Luca Vangelista
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Nur-Sultan, Kazakhstan
| | | |
Collapse
|
55
|
Failed Disruption of Tick Feeding, Viability, and Molting after Immunization of Mice and Sheep with Recombinant Ixodes ricinus Salivary Proteins IrSPI and IrLip1. Vaccines (Basel) 2020; 8:vaccines8030475. [PMID: 32858821 PMCID: PMC7564719 DOI: 10.3390/vaccines8030475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
To identify potential vaccine candidates against Ixodes ricinus and tick-borne pathogen transmission, we have previously sequenced the salivary gland transcriptomes of female ticks infected or not with Bartonella henselae. The hypothesized potential of both IrSPI (I. ricinus serine protease inhibitor) and IrLip1 (I. ricinus lipocalin 1) as protective antigens decreasing tick feeding and/or the transmission of tick-borne pathogens was based on their presumed involvement in dampening the host immune response to tick feeding. Vaccine endpoints included tick larval and nymphal mortality, feeding, and molting in mice and sheep. Whether the antigens were administered individually or in combination, the vaccination of mice or sheep elicited a potent antigen-specific antibody response. However, and contrary to our expectations, vaccination failed to afford protection against the infestation of mice and sheep by I. ricinus nymphs and larvae, respectively. Rather, vaccination with IrSPI and IrLip1 appeared to enhance tick engorgement and molting and decrease tick mortality. To the best of our knowledge, these observations represent the first report of induction of vaccine-mediated enhancement in relation to anti-tick vaccination.
Collapse
|
56
|
Kostoff RN, Briggs MB, Porter AL, Aschner M, Spandidos DA, Tsatsakis A. [Editorial] COVID‑19: Post‑lockdown guidelines. Int J Mol Med 2020; 46:463-466. [PMID: 32626934 PMCID: PMC7307834 DOI: 10.3892/ijmm.2020.4640] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
Since March, 2020, in response to the COVID‑19 pandemic, many countries have been on lockdown (at different levels of severity), restricting many activities and businesses that involve gatherings of large numbers of people in close proximity. Currently (early June, 2020), countries across the globe are in different stages of easing lockdown restrictions. Public policies for behaviors and actions during this transition period vary widely across countries and within country jurisdictions. The present editorial will address potential policies that could minimize resurgence of the present pandemic (the 'second‑wave') and reduce the likelihood and severity of similar future pandemics.
Collapse
Affiliation(s)
- Ronald N. Kostoff
- School of Public Policy, Georgia Institute of Technology, Gainesville, VA 20155
| | | | - Alan L. Porter
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA 30332
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
57
|
Zellweger RM, Wartel TA, Marks F, Song M, Kim JH. Vaccination against SARS-CoV-2 and disease enhancement - knowns and unknowns. Expert Rev Vaccines 2020; 19:691-698. [PMID: 32838605 PMCID: PMC7566857 DOI: 10.1080/14760584.2020.1800463] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/21/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The world is currently fighting a COVID-19 pandemic, perhaps the most disruptive infectious disease outbreak since the 1918 Spanish influenza. Governments have taken drastic measures to curb the spread of SARS-CoV-2, and the development of safe and efficacious vaccine candidates is being accelerated. The possibility of vaccine-mediated disease enhancement with coronavirus vaccines has been flagged as a potential safety concern, and, despite the urgent need, should be thoroughly assessed as vaccines against SARS-CoV-2 are being tested. AREA COVERED We review the in vivo evidence suggesting a theoretical risk of disease enhancement after vaccination with SARS-CoV and MERS-CoV vaccine candidates. We also identify knowledge gaps that need to be filled to maximize the chance of developing a safe vaccine and minimize the risk of encountering disease enhancement in vaccinated individuals after exposure to SARS-CoV-2. EXPERT OPINION We compile and propose avenues to investigate the risk of vaccine-mediated disease enhancement both during pre-clinical and early clinical development. While the pressing need for a vaccine against COVID-19 (and future epidemic coronaviruses) cannot be ignored, we advocate to keep safety at the center of the debate. Protecting individuals with effective and safe vaccines should be a priority, even during extraordinary times like the COVID-19 pandemic.
Collapse
Affiliation(s)
- Raphaël M. Zellweger
- Epidemiology, Public Health, Impact & Clinical Development Unit, International Vaccine Institute, Seoul, Republic of Korea
| | - T. Anh Wartel
- Epidemiology, Public Health, Impact & Clinical Development Unit, International Vaccine Institute, Seoul, Republic of Korea
| | - Florian Marks
- Epidemiology, Public Health, Impact & Clinical Development Unit, International Vaccine Institute, Seoul, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul, Republic of Korea
| | - Jerome H. Kim
- Epidemiology, Public Health, Impact & Clinical Development Unit, International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
58
|
Curlin ME, Shao J, Diaz G, Edlefsen PT, Novak RM, Mayer KH, Allen M, Morgan C, Maenza J, Buchbinder S, Keefer MC, Rosa SCD, Corey L, Duerr A. Long-term mucosal T cell activation and homing phenotypes in recipients of an Ad5-vectored HIV vaccine. Vaccine 2020; 38:5814-5821. [PMID: 32680773 DOI: 10.1016/j.vaccine.2020.06.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/06/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vaccine-induced mucosal immune responses may be critical for protection against HIV infection, but may also result in short or long-term changes that enhance susceptibility to infection in some individuals, such as those with baseline seroreactivity to vaccine vector antigens. We examined cellular immune responses in blood and gut mucosal tissue roughly two years following vaccination with placebo or the Step study vaccine MRKAd5/HIV-1. METHODS Participants vaccinated with either placebo or MRKAd5/HIV-1 during participation in HVTN 071, and HVTN 502/Merck 023 underwent phlebotomy and colonic mucosal biopsies via flexible sigmoidoscopy at two timepoints roughly six months apart. After isolation of mononuclear cells, we compared cellular phenotypes and intracellular cytokine responses in vaccine and placebo recipients with and without baseline serological reactivity to Ad5. RESULTS Surface expression of activation and gut-homing markers were elevated on CD4 + and CD8 + gut mucosal mononuclear cells (GMMC) in comparison with PBMC (p < 0.01), but were not significantly affected by baseline Ad5 serostatus or receipt of MRKAd5/HIV-1. ICS responses to stimulation with vaccine antigens were of low frequency and magnitude. Ad5 vector responses were seen in vaccinees and baseline seropositive individuals. CD4 + responses to vector antigens were more common in GMMC than PBMC (p < 0.01) and CD8 + responses to HIV gag insert antigens were more frequent in Ad5 seropositive than Ad5 seronegative individuals (p = 0.03). CONCLUSION Vaccination with the Ad5 vectored candidate HIV vaccine MRKAd5/HIV-1 does not lead to long-term changes in the activation state of mucosal CD4 + or CD8 + T lymphocytes regardless of baseline Ad5 serostatus. The findings of this study do not reveal a basis for enhanced susceptibility to HIV infection two years post vaccination.
Collapse
Affiliation(s)
- Marcel E Curlin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | - Jason Shao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Gabriela Diaz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Richard M Novak
- Division of Infectious Diseases, University of Illinois, Chicago, USA
| | - Kenneth H Mayer
- Fenway Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mary Allen
- Vaccine Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecilia Morgan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Susan Buchbinder
- Bridge HIV, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Michael C Keefer
- Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Stephen C De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ann Duerr
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | |
Collapse
|
59
|
Abstract
COVID-19 has become one of the biggest health concern, along with huge economic burden. With no clear remedies to treat the disease, doctors are repurposing drugs like chloroquine and remdesivir to treat COVID-19 patients. In parallel, research institutes in collaboration with biotech companies have identified strategies to use viral proteins as vaccine candidates for COVID-19. Although this looks promising, they still need to pass the test of challenge studies in animal models. As various models for SARS-CoV-2 are under testing phase, biotech companies have bypassed animal studies and moved to Phase I clinical trials. In view of the present outbreak, this looks a justified approach, but the problem is that in the absence of animal studies, we can never predict the outcomes in humans. Since animal models are critical for vaccine development and SARS-CoV-2 has different transmission dynamics, in this review we compare different animal models of SARS-CoV-2 with humans for their pathogenic, immune response and transmission dynamics that make them ideal models for vaccine testing for COVID-19. Another issue of using animal model is the ethics of using animals for research; thus, we also discuss the pros and cons of using animals for vaccine development studies.
Collapse
|
60
|
Lega S, Naviglio S, Volpi S, Tommasini A. Recent Insight into SARS-CoV2 Immunopathology and Rationale for Potential Treatment and Preventive Strategies in COVID-19. Vaccines (Basel) 2020; 8:E224. [PMID: 32423059 PMCID: PMC7349555 DOI: 10.3390/vaccines8020224] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
As the outbreak of the new coronavirus (SARS-CoV-2) infection is spreading globally, great effort is being made to understand the disease pathogenesis and host factors that predispose to disease progression in an attempt to find a window of opportunity for intervention. In addition to the direct cytopathic effect of the virus, the host hyper-inflammatory response has emerged as a key factor in determining disease severity and mortality. Accumulating clinical observations raised hypotheses to explain why some patients develop more severe disease while others only manifest mild or no symptoms. So far, Covid-19 management remains mainly supportive. However, many researches are underway to clarify the role of antiviral and immunomodulating drugs in changing morbidity and mortality in patients who become severely ill. This review summarizes the current state of knowledge on the interaction between SARS-CoV-2 and the host immune system and discusses recent findings on proposed pharmacologic treatments.
Collapse
Affiliation(s)
- Sara Lega
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
| | - Samuele Naviglio
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
| | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiency, IRCCS Istituto Giannina Gaslini and Università degli Studi di Genova, 16147 Genova, Italy;
| | - Alberto Tommasini
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34137 Trieste, Italy
| |
Collapse
|
61
|
Infection-Immunity Competition: A Simple Model for Illustrating the Background of Individual Response on Herd Immunity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10093078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
For achieving herd immunity, the proportion of individuals who are immunized, and the proportion of susceptible individuals are normally regarded as the key factors. Here, it is discussed that the immunity is not a yes/no decision in all cases, but a limited (relative) immunity should be kept in mind. This effect would cause a dependence of infection from the level of immunity and the strength of single-infection impact events (virus load). As a result, a stepwise enhancement of low-level immunity could be achieved in case of infection contacts at low concentrations of infectious particles. This behavior is probably important for airborne infection paths. Therefore, it might play a role in the case of the recent SARS (new coronavirus) pandemic and could have a strong effect on herd immunity.
Collapse
|
62
|
|
63
|
Association of complement C3d receptor 2 genotypes with the acquisition of HIV infection in a trial of recombinant glycoprotein 120 vaccine. AIDS 2020; 34:25-32. [PMID: 31634193 DOI: 10.1097/qad.0000000000002401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Complement C3d receptor 2 (CR2) is the main receptor for complement protein C3d and plays an important role in adaptive immune responses. CR2 genetic variants are associated with susceptibility to systemic lupus erythematosus as well as to HIV-1 infection. In addition, CR2 function can be subverted by HIV-1 for an efficient entry into target cells; in a process known as antibody-dependent enhancement of viral infection. We sought to determine the association between CR2 gene variants with HIV-1 acquisition after vaccination with recombinant gp120 protein (Vax004 clinical trial). DESIGN AND METHODS This is a retrospective cross-sectional study, comprising male volunteers of European ancestry including infected (n = 273) and uninfected (n = 402) vaccinees and placebo, who were genotyped for three single nucleotide polymorphisms (SNPs) in the CR2 gene region. RESULTS An interaction was observed between the baseline sexual behavior and the SNP rs3813946 for higher risk of infection in vacinees (interaction term P = 0.02). This SNP was associated with increased susceptibility to HIV-1 infection after vaccination in volunteers with low behavioral risk odds ratio (95% confidence interval): 5.5 (1.4-21.7) P = 0.006 but not vaccinees with high behavioral risk or volunteers given placebo (P = 0.7). Moreover, CR2 genotype was strongly associated with the rate of HIV-1 acquisition after vaccination in low-risk volunteers [hazard odds ratio (95% confidence interval): 3.3 (1.6-7.0), P = 0.001]. CONCLUSION The current study suggests that CR2 may play a role in HIV-1 acquisition after vaccination with rgp120 proteins.
Collapse
|
64
|
Abdala A, Alvarez I, Brossel H, Calvinho L, Carignano H, Franco L, Gazon H, Gillissen C, Hamaidia M, Hoyos C, Jacques JR, Joris T, Laval F, Petersen M, Porquet F, Porta N, Ruiz V, Safari R, Suárez Archilla G, Trono K, Willems L. BLV: lessons on vaccine development. Retrovirology 2019; 16:26. [PMID: 31590667 PMCID: PMC6781361 DOI: 10.1186/s12977-019-0488-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/10/2019] [Indexed: 01/12/2023] Open
Abstract
Vaccination against retroviruses is a challenge because of their ability to stably integrate into the host genome, undergo long-term latency in a proportion of infected cells and thereby escape immune response. Since clearance of the virus is almost impossible once infection is established, the primary goal is to achieve sterilizing immunity. Besides efficacy, safety is the major issue since vaccination has been associated with increased infection or reversion to pathogenicity. In this review, we discuss the different issues that we faced during the development of an efficient vaccine against bovine leukemia virus (BLV). We summarize the historical failures of inactivated vaccines, the efficacy and safety of a live-attenuated vaccine and the economical constraints of further industrial development.
Collapse
Affiliation(s)
- Alejandro Abdala
- Estacion Experimental Agropecuaria Rafaela, INTA, 2300, Rafaela, Argentina
| | - Irene Alvarez
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Hélène Brossel
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Luis Calvinho
- Estacion Experimental Agropecuaria Rafaela, INTA, 2300, Rafaela, Argentina
| | - Hugo Carignano
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Lautaro Franco
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Hélène Gazon
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Christelle Gillissen
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Malik Hamaidia
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Clotilde Hoyos
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Jean-Rock Jacques
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Thomas Joris
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Florent Laval
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Marcos Petersen
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Florent Porquet
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Natalia Porta
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Vanesa Ruiz
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Roghaiyeh Safari
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | | | - Karina Trono
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Luc Willems
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium. .,Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA) of University of Liège (ULiège), B34, 1 avenue de l'Hôpital, Sart-Tilman, 4000, Liege, Belgium.
| |
Collapse
|
65
|
Zuo Y, Whitbeck JC, Haila GJ, Hakim AA, Rothlauf PW, Eisenberg RJ, Cohen GH, Krummenacher C. Saliva enhances infection of gingival fibroblasts by herpes simplex virus 1. PLoS One 2019; 14:e0223299. [PMID: 31581238 PMCID: PMC6776388 DOI: 10.1371/journal.pone.0223299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 09/19/2019] [Indexed: 02/01/2023] Open
Abstract
Oral herpes is a highly prevalent infection caused by herpes simplex virus 1 (HSV-1). After an initial infection of the oral cavity, HSV-1 remains latent in sensory neurons of the trigeminal ganglia. Episodic reactivation of the virus leads to the formation of mucocutaneous lesions (cold sores), but asymptomatic reactivation accompanied by viral shedding is more frequent and allows virus spread to new hosts. HSV-1 DNA has been detected in many oral tissues. In particular, HSV-1 can be found in periodontal lesions and several studies associated its presence with more severe periodontitis pathologies. Since gingival fibroblasts may become exposed to salivary components in periodontitis lesions, we analyzed the effect of saliva on HSV-1 and -2 infection of these cells. We observed that human gingival fibroblasts can be infected by HSV-1. However, pre-treatment of these cells with saliva extracts from some but not all individuals led to an increased susceptibility to infection. Furthermore, the active saliva could expand HSV-1 tropism to cells that are normally resistant to infection due to the absence of HSV entry receptors. The active factor in saliva was partially purified and comprised high molecular weight complexes of glycoproteins that included secretory Immunoglobulin A. Interestingly, we observed a broad variation in the activity of saliva between donors suggesting that this activity is selectively present in the population. The active saliva factor, has not been isolated, but may lead to the identification of a relevant biomarker for susceptibility to oral herpes. The presence of a salivary factor that enhances HSV-1 infection may influence the risk of oral herpes and/or the severity of associated oral pathologies.
Collapse
Affiliation(s)
- Yi Zuo
- Department of Microbiology, School of Dental Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - J. Charles Whitbeck
- Department of Microbiology, School of Dental Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gabriel J. Haila
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, United States of America
| | - Abraham A. Hakim
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
| | - Paul W. Rothlauf
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
| | - Roselyn J. Eisenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gary H. Cohen
- Department of Microbiology, School of Dental Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Claude Krummenacher
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, New Jersey, United States of America
| |
Collapse
|
66
|
|
67
|
Lokhandwala S, Petrovan V, Popescu L, Sangewar N, Elijah C, Stoian A, Olcha M, Ennen L, Bray J, Bishop RP, Waghela SD, Sheahan M, Rowland RRR, Mwangi W. Adenovirus-vectored African Swine Fever Virus antigen cocktails are immunogenic but not protective against intranasal challenge with Georgia 2007/1 isolate. Vet Microbiol 2019; 235:10-20. [PMID: 31282366 DOI: 10.1016/j.vetmic.2019.06.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 11/29/2022]
Abstract
African Swine Fever Virus (ASFV) causes a hemorrhagic disease in swine and wild boars with a fatality rate close to 100%. Less virulent strains cause subchronic or chronic forms of the disease. The virus is endemic in sub-Saharan Africa and an outbreak in Georgia in 2007 spread to Armenia, Russia, Ukraine, Belarus, Poland, Lithuania, and Latvia. In August 2018, there was an outbreak in China and in April 2019, ASFV was reported in Vietnam and Cambodia. Since no vaccine or treatment exists, a vaccine is needed to safeguard the swine industry. Previously, we evaluated immunogenicity of two adenovirus-vectored cocktails containing ASFV antigens and demonstrated induction of unprecedented robust antibody and T cell responses, including cytotoxic T lymphocytes. In the present study, we evaluated protective efficacy of both cocktails by intranasal challenge of pigs with ASFV-Georgia 2007/1. A nine antigen cocktail-(I) formulated in BioMize adjuvant induced strong IgG responses, but when challenged, the vaccinees had more severe reaction relative to the controls. A seven antigen cocktail-(II) was evaluated using two adjuvants: BioMize and ZTS-01. The BioMize formulation induced stronger antibody responses, but 8/10 vaccinees and 4/5 controls succumbed to the disease or reached experimental endpoint at 17 days post-challenge. In contrast, the ZTS-01 formulation induced weaker antibody responses, but 4/9 pigs succumbed to the disease while the 5 survivors exhibited low clinical scores and no viremia at 17 days post-challenge, whereas 4/5 controls succumbed to the disease or reached experimental endpoint. Overall, none of the immunogens conferred statistically significant protection.
Collapse
Affiliation(s)
- Shehnaz Lokhandwala
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Vlad Petrovan
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Luca Popescu
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Neha Sangewar
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Catherine Elijah
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Ana Stoian
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Matthew Olcha
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Lindsey Ennen
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Jocelyn Bray
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Richard P Bishop
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman WA, United States
| | - Suryakant D Waghela
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Maureen Sheahan
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Raymond R R Rowland
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Waithaka Mwangi
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
68
|
Post-exposure administration of chimeric antibody protects mice against European, Siberian, and Far-Eastern subtypes of tick-borne encephalitis virus. PLoS One 2019; 14:e0215075. [PMID: 30958863 PMCID: PMC6453444 DOI: 10.1371/journal.pone.0215075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/26/2019] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is the most important tick-transmitted pathogen. It belongs to the Flaviviridae family and causes severe human neuroinfections. In this study, protective efficacy of the chimeric antibody chFVN145 was examined in mice infected with strains belonging to the Far-Eastern, European, and Siberian subtypes of TBEV, and the antibody showed clear therapeutic efficacy when it was administered once one, two, or three days after infection. The efficacy was independent of the TBEV strain used to infect the mice; however, the survival rate of the mice was dependent on the dose of TBEV and of the antibody. No enhancement of TBEV infection was observed when the mice were treated with non-protective doses of chFVN145. Using a panel of recombinant fragments of the TBEV glycoprotein E, the neutralizing epitope for chFVN145 was localized in domain III of the TBEV glycoprotein E, in a region between amino acid residues 301 and 359. In addition, three potential sites responsible for binding with chFVN145 were determined using peptide phage display libraries, and 3D modeling demonstrated that the sites do not contact the fusion loop and, hence, their binding with chFVN145 does not result in increased attachment of TBEV to target cells.
Collapse
|
69
|
Tick-borne encephalitis in Europe and Russia: Review of pathogenesis, clinical features, therapy, and vaccines. Antiviral Res 2019; 164:23-51. [PMID: 30710567 DOI: 10.1016/j.antiviral.2019.01.014] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/10/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023]
Abstract
Tick-borne encephalitis (TBE) is an illness caused by tick-borne encephalitis virus (TBEV) infection which is often limited to a febrile illness, but may lead to very aggressive downstream neurological manifestations. The disease is prevalent in forested areas of Europe and northeastern Asia, and is typically caused by infection involving one of three TBEV subtypes, namely the European (TBEV-Eu), the Siberian (TBEV-Sib), or the Far Eastern (TBEV-FE) subtypes. In addition to the three main TBEV subtypes, two other subtypes; i.e., the Baikalian (TBEV-Bkl) and the Himalayan subtype (TBEV-Him), have been described recently. In Europe, TBEV-Eu infection usually results in only mild TBE associated with a mortality rate of <2%. TBEV-Sib infection also results in a generally mild TBE associated with a non-paralytic febrile form of encephalitis, although there is a tendency towards persistent TBE caused by chronic viral infection. TBE-FE infection is considered to induce the most severe forms of TBE. Importantly though, viral subtype is not the sole determinant of TBE severity; both mild and severe cases of TBE are in fact associated with infection by any of the subtypes. In keeping with this observation, the overall TBE mortality rate in Russia is ∼2%, in spite of the fact that TBEV-Sib and TBEV-FE subtypes appear to be inducers of more severe TBE than TBEV-Eu. On the other hand, TBEV-Sib and TBEV-FE subtype infections in Russia are associated with essentially unique forms of TBE rarely seen elsewhere if at all, such as the hemorrhagic and chronic (progressive) forms of the disease. For post-exposure prophylaxis and TBE treatment in Russia and Kazakhstan, a specific anti-TBEV immunoglobulin is currently used with well-documented efficacy, but the use of specific TBEV immunoglobulins has been discontinued in Europe due to concerns regarding antibody-enhanced disease in naïve individuals. Therefore, new treatments are essential. This review summarizes available data on the pathogenesis and clinical features of TBE, plus different vaccine preparations available in Europe and Russia. In addition, new treatment possibilities, including small molecule drugs and experimental immunotherapies are reviewed. The authors caution that their descriptions of approved or experimental therapies should not be considered to be recommendations for patient care.
Collapse
|
70
|
Smatti MK, Al Thani AA, Yassine HM. Viral-Induced Enhanced Disease Illness. Front Microbiol 2018; 9:2991. [PMID: 30568643 PMCID: PMC6290032 DOI: 10.3389/fmicb.2018.02991] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding immune responses to viral infections is crucial to progress in the quest for effective infection prevention and control. The host immunity involves various mechanisms to combat viral infections. Under certain circumstances, a viral infection or vaccination may result in a subverted immune system, which may lead to an exacerbated illness. Clinical evidence of enhanced illness by preexisting antibodies from vaccination, infection or maternal passive immunity is available for several viruses and is presumptively proposed for other viruses. Multiple mechanisms have been proposed to explain this phenomenon. It has been confirmed that certain infection- and/or vaccine-induced immunity could exacerbate viral infectivity in Fc receptor- or complement bearing cells- mediated mechanisms. Considering that antibody dependent enhancement (ADE) is a major obstacle in vaccine development, there are continues efforts to understand the underlying mechanisms through identification of the epitopes and antibodies responsible for disease enhancement or protection. This review discusses the recent findings on virally induced ADE, and highlights the potential mechanisms leading to this condition.
Collapse
Affiliation(s)
- Maria K Smatti
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
71
|
FIV vaccine with receptor epitopes results in neutralizing antibodies but does not confer resistance to challenge. NPJ Vaccines 2018; 3:16. [PMID: 29736270 PMCID: PMC5928050 DOI: 10.1038/s41541-018-0051-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 11/09/2022] Open
Abstract
Feline immunodeficiency virus (FIV) is the feline analogue to human immunodeficiency virus (HIV) and utilizes parallel modes of receptor-mediated entry. The FIV surface glycoprotein (SU) is an important target for induction of neutralizing antibodies, and autoantibodies to the FIV binding receptor (CD134) block infection ex vivo; thus highlighting the potential for immunotherapies which utilize anti-receptor antibodies to block viral infection. To determine whether vaccination with CD134-SU complexes could induce protection against FIV infection, cats (n = 5 per group) were immunized with soluble CD134, recombinant FIV-SU protein, and/or CD134+SU complexes. Two trials were performed with different antigen combinations and vaccination schedules. In vivo generation of anti-CD134 and anti-SU IgG antibodies was measured, and in vitro neutralization assays were conducted. Immunization induced production of anti-CD134 and anti-SU antibodies that significantly inhibited FIV infection in vitro. However, no vaccine combination protected cats from FIV infection, and neat serum from vaccinated cats enhanced FIV growth in vitro. CD134+SU vaccinated cats exhibited increased CD4:CD8 ratio immediately prior to challenge, and antibodies were much more efficiently generated against vaccine by-products versus target antigens. Results suggest vaccination against viral and cryptic receptor epitopes yields neutralizing antibodies that synergistically inhibit FIV infection in vitro. Factors contributing to vaccine failure may include: (1) Heat-labile serum factors that enhance viral replication, (2) changes in circulating target cell populations induced by vaccination, and (3) weak immunogenicity of neutralizing epitopes compared to off-target vaccine components. Results reinforce the need to monitor vaccine preparation components and avoid non-specific immune stimulation during vaccination. A vaccine candidate for feline immunodeficiency virus elicits strong immunological reaction in vitro, but no protection to live cats. The feline analog to human immunodeficiency virus, FIV shares a similar infection paradigm and has only one partially effective vaccine. A US team, led by Colorado State University’s Susan VandeWoude, immunized cats using a complex of an FIV surface protein and a feline cell-surface protein known to facilitate FIV’s entry into immune cells. Tissue culture assays yielded promising results; however, this did not translate to live-animal protection. The researchers highlighted multiple factors that could explain the lack of success, including circulatory pro-infection factors, and immune responses generated against vaccine by-products rather than intended targets. While the vaccine candidate failed, the research provides invaluable guidance for future efforts into FIV vaccination with implications for HIV vaccine trials.
Collapse
|
72
|
Applications of the FIV Model to Study HIV Pathogenesis. Viruses 2018; 10:v10040206. [PMID: 29677122 PMCID: PMC5923500 DOI: 10.3390/v10040206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Feline immunodeficiency virus (FIV) is a naturally-occurring retrovirus that infects domestic and non-domestic feline species, producing progressive immune depletion that results in an acquired immunodeficiency syndrome (AIDS). Much has been learned about FIV since it was first described in 1987, particularly in regard to its application as a model to study the closely related lentivirus, human immunodeficiency virus (HIV). In particular, FIV and HIV share remarkable structure and sequence organization, utilize parallel modes of receptor-mediated entry, and result in a similar spectrum of immunodeficiency-related diseases due to analogous modes of immune dysfunction. This review summarizes current knowledge of FIV infection kinetics and the mechanisms of immune dysfunction in relation to opportunistic disease, specifically in regard to studying HIV pathogenesis. Furthermore, we present data that highlight changes in the oral microbiota and oral immune system during FIV infection, and outline the potential for the feline model of oral AIDS manifestations to elucidate pathogenic mechanisms of HIV-induced oral disease. Finally, we discuss advances in molecular biology, vaccine development, neurologic dysfunction, and the ability to apply pharmacologic interventions and sophisticated imaging technologies to study experimental and naturally occurring FIV, which provide an excellent, but often overlooked, resource for advancing therapies and the management of HIV/AIDS.
Collapse
|
73
|
Development of Antibody Therapeutics against Flaviviruses. Int J Mol Sci 2017; 19:ijms19010054. [PMID: 29295568 PMCID: PMC5796004 DOI: 10.3390/ijms19010054] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/28/2022] Open
Abstract
Recent outbreaks of Zika virus (ZIKV) highlight the urgent need to develop efficacious interventions against flaviviruses, many of which cause devastating epidemics around the world. Monoclonal antibodies (mAb) have been at the forefront of treatment for cancer and a wide array of other diseases due to their specificity and potency. While mammalian cell-produced mAbs have shown promise as therapeutic candidates against several flaviviruses, their eventual approval for human application still faces several challenges including their potential risk of predisposing treated patients to more severe secondary infection by a heterologous flavivirus through antibody-dependent enhancement (ADE). The high cost associated with mAb production in mammalian cell cultures also poses a challenge for the feasible application of these drugs to the developing world where the majority of flavivirus infection occurs. Here, we review the current therapeutic mAb candidates against various flaviviruses including West Nile (WNV), Dengue virus (DENV), and ZIKV. The progress of using plants for developing safer and more economical mAb therapeutics against flaviviruses is discussed within the context of their expression, characterization, downstream processing, neutralization, and in vivo efficacy. The progress of using plant glycoengineering to address ADE, the major impediment of flavivirus therapeutic development, is highlighted. These advancements suggest that plant-based systems are excellent alternatives for addressing the remaining challenges of mAb therapeutic development against flavivirus and may facilitate the eventual commercialization of these drug candidates.
Collapse
|
74
|
Palermo A, Weber LK, Rentschler S, Isse A, Sedlmayr M, Herbster K, List V, Hubbuch J, Löffler FF, Nesterov-Müller A, Breitling F. Identification of a Tetanus Toxin Specific Epitope in Single Amino Acid Resolution. Biotechnol J 2017; 12. [PMID: 28922578 DOI: 10.1002/biot.201700197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/14/2017] [Indexed: 01/24/2023]
Abstract
Vaccinations are among the most potent tools to fight infectious diseases. However, cross-reactions are an ongoing problem and there is an urgent need to fully understand the mechanisms of the immune response. For the development of a methodological workflow, the linear epitopes in the immune response to the tetanus toxin is investigated in sera of 19 vaccinated Europeans applying epitope mapping with peptide arrays. The most prominent epitope, appearing in nine different sera (923 IHLVNNESSEVIVHK937 ), is investigated in a substitution analysis to identify the amino acids that are crucial for the binding of the corresponding antibody species - the antibody fingerprint. The antibody fingerprints of different individuals are compared and found to be strongly conserved (929 ExxEVIVxK937 ), which is astonishing considering the randomness of their development. Additionally, the corresponding antibody species is isolated from one serum with batch chromatography using the amino acid sequence of the identified epitope and the tetanus specificity of the isolated antibody is verified by ELISA. Studying antibody fingerprints with peptide arrays should be transferable to any kind of humoral immune response toward protein antigens. Furthermore, antibody fingerprints have shown to be highly disease-specific and, therefore, can be employed as reliable biomarkers enabling the study of cross-reacting antigens.
Collapse
Affiliation(s)
- Andrea Palermo
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Laura K Weber
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Simone Rentschler
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Awale Isse
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Martyna Sedlmayr
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Karin Herbster
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Volker List
- Medical Services, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Jürgen Hubbuch
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Felix F Löffler
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Alexander Nesterov-Müller
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| | - Frank Breitling
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Karlsruhe, Germany
| |
Collapse
|
75
|
Lotrič-Furlan S, Bogovič P, Avšič-Županc T, Jelovšek M, Lusa L, Strle F. Tick-borne encephalitis in patients vaccinated against this disease. J Intern Med 2017; 282:142-155. [PMID: 28440879 DOI: 10.1111/joim.12625] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Information on tick-borne encephalitis (TBE) in patients already vaccinated against the disease is limited. OBJECTIVES To compare the course and outcome in patients with vaccination breakthrough TBE with findings in patients who developed TBE without previous vaccination. METHODS All adult patients diagnosed with TBE at a single medical centre during a 16-year period and who had received at least two doses of TBE vaccine before the onset of illness qualified for the study. For each patient with breakthrough TBE, two unvaccinated sex- and age-matched patients, diagnosed with TBE in the same year, were included for comparison. RESULTS Amongst 2332 patients diagnosed with TBE in the period 2000-2015, 39 (1.7%) had been vaccinated against the disease. Their median age was 59 (20-83) years; 22 of 39 (56.4%) were male. In comparison with unvaccinated patients with TBE, those with breakthrough disease more often experienced a monophasic course of illness (P = 0.006), had a higher CSF leucocyte count (P = 0.005), more often had urine retention (P = 0.012), more often needed ICU treatment (P = 0.009), were hospitalized for longer (P = 0.002) and had more severe acute illness (P = 0.004 for simple clinical assessment, P = 0.001 for severity score). CONCLUSION In addition to several findings corroborating previous results in patients with vaccination breakthrough TBE, such as older age and the presence of a particular specific serum antibody pattern indicating anamnestic response, findings in this study indicate that the acute illness in patients with breakthrough TBE is more severe than in unvaccinated sex- and age-matched patients who develop the disease.
Collapse
Affiliation(s)
- S Lotrič-Furlan
- Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - P Bogovič
- Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - T Avšič-Županc
- Medical Faculty, Institute for Microbiology and Immunology, Ljubljana, Slovenia
| | - M Jelovšek
- Medical Faculty, Institute for Microbiology and Immunology, Ljubljana, Slovenia
| | - L Lusa
- Faculty of Medicine, Institute for Biostatistics and Medical Informatics, University of Ljubljana, Ljubljana, Slovenia
| | - F Strle
- Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
76
|
Yamanaka A, Moi ML, Takasaki T, Kurane I, Konishi E. Neutralizing and enhancing antibody responses to five genotypes of dengue virus type 1 (DENV-1) in DENV-1 patients. J Gen Virol 2017; 98:166-172. [PMID: 27911254 DOI: 10.1099/jgv.0.000669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV) has four distinct serotypes, DENV-1-4, with four to six genotypes in each serotype. The World Health Organization recommends tetravalent formulations including one genotype of each serotype as safe and effective dengue vaccines. Here, we investigated the impact of genotype on the neutralizing antibody responses to DENV-1 in humans. Convalescent sera collected from patients with primary infection of DENV-1 were examined for neutralizing antibody against single-round infectious particles of the five DENV-1 genotypes (GI-GV). In both GI- and GIV-infected patients, their neutralizing antibody titres against the five genotypes were similar, differing ≤4-fold from the homogenotypic responses. The enhancing activities against the five genotypes were also similar in these sera. Thus, the genotype strains of DENV-1 showed no significant antigenic differences in these patients, suggesting that GI- or GIV-derived vaccine antigens should induce equivalent levels of neutralizing antibodies against all DENV-1 genotypes.
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand.,BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Meng Ling Moi
- Present address: Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomohiko Takasaki
- Present address: Kanagawa Prefectural Institute of Public Health, Kanagawa, Japan.,Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Ichiro Kurane
- National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.,BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| |
Collapse
|
77
|
Dent M, Hurtado J, Paul AM, Sun H, Lai H, Yang M, Esqueda A, Bai F, Steinkellner H, Chen Q. Plant-produced anti-dengue virus monoclonal antibodies exhibit reduced antibody-dependent enhancement of infection activity. J Gen Virol 2016; 97:3280-3290. [PMID: 27902333 PMCID: PMC5756494 DOI: 10.1099/jgv.0.000635] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
The mAb E60 has the potential to be a desirable therapeutic molecule since it efficiently neutralizes all four serotypes of dengue virus (DENV). However, mammalian-cell-produced E60 exhibits antibody-dependent enhancement of infection (ADE) activity, rendering it inefficacious in vivo, and treated animals more susceptible to developing more severe diseases during secondary infection. In this study, we evaluated a plant-based expression system for the production of therapeutically suitable E60. The mAb was transiently expressed in Nicotiana benthamianaWT and a ∆XFT line, a glycosylation mutant lacking plant-specific N-glycan residues. The mAb was efficiently expressed and assembled in leaves and exhibited highly homogenous N-glycosylation profiles, i.e. GnGnXF3 or GnGn structures, depending on the expression host. Both E60 glycovariants demonstrated equivalent antigen-binding specificity and in vitro neutralization potency against DENV serotypes 2 and 4 compared with their mammalian-cell-produced counterpart. By contrast, plant-produced E60 exhibited reduced ADE activity in Fc gamma receptor expressing human cells. Our results suggest the ability of plant-produced antibodies to minimize ADE, which may lead to the development of safe and highly efficacious antibody-based therapeutics against DENV and other ADE-prone viral diseases. Our study provides so far unknown insight into the relationship between mAb N-glycosylation and ADE, which contributes to our understanding of how sugar moieties of antibodies modulate Fc-mediated functions and viral pathogenesis.
Collapse
Affiliation(s)
- Matthew Dent
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jonathan Hurtado
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Amber M. Paul
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS, USA
| | - Haiyan Sun
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Huafang Lai
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Ming Yang
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Adrian Esqueda
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Fengwei Bai
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS, USA
| | - Herta Steinkellner
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Qiang Chen
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
78
|
Taylor A, Foo SS, Bruzzone R, Dinh LV, King NJC, Mahalingam S. Fc receptors in antibody-dependent enhancement of viral infections. Immunol Rev 2016; 268:340-64. [PMID: 26497532 PMCID: PMC7165974 DOI: 10.1111/imr.12367] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sensitization of the humoral immune response to invading viruses and production of antiviral antibodies forms part of the host antiviral repertoire. Paradoxically, for a number of viral pathogens, under certain conditions, antibodies provide an attractive means of enhanced virus entry and replication in a number of cell types. Known as antibody‐dependent enhancement (ADE) of infection, the phenomenon occurs when virus‐antibody immunocomplexes interact with cells bearing complement or Fc receptors, promoting internalization of the virus and increasing infection. Frequently associated with exacerbation of viral disease, ADE of infection presents a major obstacle to the prevention of viral disease by vaccination and is thought to be partly responsible for the adverse effects of novel antiviral therapeutics such as intravenous immunoglobulins. There is a growing body of work examining the intracellular signaling pathways and epitopes responsible for mediating ADE, with a view to aiding rational design of antiviral strategies. With in vitro studies also confirming ADE as a feature of infection for a growing number of viruses, challenges remain in understanding the multilayered molecular mechanisms of ADE and its effect on viral pathogenesis.
Collapse
Affiliation(s)
- Adam Taylor
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Suan-Sin Foo
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong SAR, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Luan Vu Dinh
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Nicholas J C King
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Suresh Mahalingam
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| |
Collapse
|
79
|
Abstract
Traditional vaccination with whole pathogens or pathogen-derived subunits has completely eliminated diseases like smallpox, and has greatly limited the incidence, morbidity and mortality associated with many other infectious diseases. Unfortunately, a large burden of infectious disease remains that may be preventable through vaccination. For many of these, more focused and innovative approaches may be essential for the development of effective vaccines.
Collapse
Affiliation(s)
- Jon Oscherwitz
- a Division of Hematology-Oncology, Department of Internal Medicine , University of Michigan Medical School , Ann Arbor , MI , USA.,b Veterans Administration Ann Arbor Healthcare System , Ann Arbor , MI , USA
| |
Collapse
|
80
|
Rajao DS, Sandbulte MR, Gauger PC, Kitikoon P, Platt R, Roth JA, Perez DR, Loving CL, Vincent AL. Heterologous challenge in the presence of maternally-derived antibodies results in vaccine-associated enhanced respiratory disease in weaned piglets. Virology 2016; 491:79-88. [PMID: 26874588 DOI: 10.1016/j.virol.2016.01.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/17/2015] [Accepted: 01/22/2016] [Indexed: 12/19/2022]
Abstract
Control of influenza A virus (IAV) in pigs is done by vaccination of females to provide maternally-derived antibodies (MDA) through colostrum. Our aim was to evaluate if MDA interfere with IAV infection, clinical disease, and transmission in non-vaccinated piglets. In the first study, naïve sows were vaccinated with H1N2-δ1 whole inactivated virus (WIV) vaccine. In a follow-up study seropositive sows to 2009 pandemic H1N1 (H1N1pdm09) were boosted with H1N1pdm09 WIV or secondary experimental infection (EXP). MDA-positive pigs were challenged with homologous or heterologous virus, and MDA-negative control groups were included. WIV-MDA piglets were protected from homologous infection. However, piglets with WIV-derived MDA subsequently challenged with heterologous virus developed vaccine associated enhanced respiratory disease (VAERD), regardless of history of natural exposure in the sows. Our data indicates that although high titers of vaccine-derived MDA reduced homologous virus infection, transmission, and disease, MDA alone was sufficient to induce VAERD upon heterologous infection.
Collapse
Affiliation(s)
- Daniela S Rajao
- Virus and Prion Research Unit, National Animal Disease Center, USDA, ARS, 1920 Dayton Ave, Ames, IA 50010, United States
| | - Matthew R Sandbulte
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 2180 Veterinary Medicine, Ames, IA 50011, United States
| | - Phillip C Gauger
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, 2630 Veterinary Medicine, Ames, IA 50011, United States
| | - Pravina Kitikoon
- Virus and Prion Research Unit, National Animal Disease Center, USDA, ARS, 1920 Dayton Ave, Ames, IA 50010, United States
| | - Ratree Platt
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 2180 Veterinary Medicine, Ames, IA 50011, United States
| | - James A Roth
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 2180 Veterinary Medicine, Ames, IA 50011, United States
| | - Daniel R Perez
- Poultry Diagnostic and Research Center, University of Georgia, 953 College Station Rd, Athens, GA 30602, United States
| | - Crystal L Loving
- Virus and Prion Research Unit, National Animal Disease Center, USDA, ARS, 1920 Dayton Ave, Ames, IA 50010, United States
| | - Amy L Vincent
- Virus and Prion Research Unit, National Animal Disease Center, USDA, ARS, 1920 Dayton Ave, Ames, IA 50010, United States.
| |
Collapse
|
81
|
Abstract
Dengue provides the most abundant example in human medicine and the greatest human illness burden caused by the phenomenon of intrinsic antibody-dependent infection enhancement (iADE). In this immunopathological phenomenon infection of monocytes or macrophages using infectious immune complexes suppresses innate antiviral systems, permitting logarithmic intracellular growth of dengue virus. The four dengue viruses evolved from a common ancestor yet retain similar ecology and pathogenicity, but although infection with one virus provides short-term cross-protection against infection with a different type, millions of secondary dengue infections occur worldwide each year. When individuals are infected in the virtual absence of cross-protective dengue antibodies, the dengue vascular permeability syndrome (DVPS) may ensue. This occurs in around 2 to 4% of second heterotypic dengue infections. A complete understanding of the biologic mechanism of iADE, dengue biology, and the mechanism of host responses to dengue infection should lead to a comprehensive and complete understanding of the pathogenesis of DVPS. A crucial emphasis must be placed on understanding ADE. Clinical and epidemiological observations of DVPS define the research questions and provide research parameters. This article will review knowledge related to dengue ADE and point to areas where there has been little research progress. These observations relate to the two stages of dengue illnesses: afferent phenomena are those that promote the success of the microorganism to infect and survive; efferent phenomena are those mounted by the host to inhibit infection and replication and to eliminate the infectious agent and infected tissues. Data will be discussed as "knowns" and "unknowns."
Collapse
|
82
|
Expression of enhancing-activity-free neutralizing antibody against dengue type 1 virus in plasmid-inoculated mice. Vaccine 2015; 33:6070-7. [DOI: 10.1016/j.vaccine.2015.07.089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/16/2015] [Accepted: 07/23/2015] [Indexed: 01/10/2023]
|
83
|
Bello-Gil D, Manez R. Exploiting natural anti-carbohydrate antibodies for therapeutic purposes. BIOCHEMISTRY (MOSCOW) 2015; 80:836-45. [DOI: 10.1134/s0006297915070044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
84
|
Gu W, Guo L, Yu H, Niu J, Huang M, Luo X, Li R, Tian Z, Feng L, Wang Y. Involvement of CD16 in antibody-dependent enhancement of porcine reproductive and respiratory syndrome virus infection. J Gen Virol 2015; 96:1712-22. [PMID: 25752917 DOI: 10.1099/vir.0.000118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The immunological effect of porcine reproductive and respiratory syndrome disease virus (PRRSV) vaccines is thought to be influenced by a variety of host factors, in which antibody-dependent enhancement (ADE) of infection is one crucial factor. Here, we assessed the mechanism of ADE of PRRSV infection. First, we found that subneutralizing serum could induce ADE of PRRSV infection in porcine alveolar macrophages (PAMs). Quantitative PCR, Western blotting and flow cytometry revealed that CD16 is the most abundant Fcγ receptor (FcγR) expressed on the surface of PAMs; thus, the role of CD16 in ADE of PRRSV infection was examined in PAMs. By using functional blocking antibodies, we demonstrated that CD16 is involved in enhanced virus production in PRRSV-antibody immune complex-infected PAMs. Because PAMs co-express different FcγR isoforms, we evaluated the effects of CD16 in FcγR-non-bearing cells by transfection. Using these engineered cells, we found that CD16 could specifically bind to the PRRSV-antibody immune complex and subsequently mediate internalization of the virus, resulting in the generation of progeny virus. We also showed that efficient expression of CD16 required association of the FcR γ-chain. Together, our findings provide significant new insights into PRRSV infection, which can be enhanced by CD16-mediated PRRSV-antibody immune complexes. This CD16-mediated ADE may induce a shift in PRRSV tropism towards CD16-expressing cells, distributing virus to more organs during virus infection.
Collapse
Affiliation(s)
- Weihong Gu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Longjun Guo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Haidong Yu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Junwei Niu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Mingming Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Xiaolei Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Ren Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Zhijun Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yue Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| |
Collapse
|
85
|
Sandbulte MR, Spickler AR, Zaabel PK, Roth JA. Optimal Use of Vaccines for Control of Influenza A Virus in Swine. Vaccines (Basel) 2015; 3:22-73. [PMID: 26344946 PMCID: PMC4494241 DOI: 10.3390/vaccines3010022] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/09/2015] [Accepted: 01/19/2015] [Indexed: 12/29/2022] Open
Abstract
Influenza A virus in swine (IAV-S) is one of the most important infectious disease agents of swine in North America. In addition to the economic burden of IAV-S to the swine industry, the zoonotic potential of IAV-S sometimes leads to serious public health concerns. Adjuvanted, inactivated vaccines have been licensed in the United States for over 20 years, and there is also widespread usage of autogenous/custom IAV-S vaccines. Vaccination induces neutralizing antibodies and protection against infection with very similar strains. However, IAV-S strains are so diverse and prone to mutation that these vaccines often have disappointing efficacy in the field. This scientific review was developed to help veterinarians and others to identify the best available IAV-S vaccine for a particular infected herd. We describe key principles of IAV-S structure and replication, protective immunity, currently available vaccines, and vaccine technologies that show promise for the future. We discuss strategies to optimize the use of available IAV-S vaccines, based on information gathered from modern diagnostics and surveillance programs. Improvements in IAV-S immunization strategies, in both the short term and long term, will benefit swine health and productivity and potentially reduce risks to public health.
Collapse
Affiliation(s)
- Matthew R Sandbulte
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Anna R Spickler
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Pamela K Zaabel
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - James A Roth
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
86
|
Shmelkov E, Nadas A, Cardozo T. Could vaccination with AIDSVAX immunogens have resulted in antibody-dependent enhancement of HIV infection in human subjects? Hum Vaccin Immunother 2014; 10:3013-6. [PMID: 25483466 DOI: 10.4161/21645515.2014.972148] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The immune-correlate analysis of the RV144 clinical trial revealed that human plasma IgA immune responses elicited by the RV144 vaccine correlated positively with a risk for HIV acquisition. This result once again emphasized that HIV vaccines can potentially have adverse effects leading to enhancement of infection. Here, we discuss previously reported evidence of antibody-dependent enhancement of HIV infection. We also describe how a structure-based epitope-specific sieve-analysis can be employed to mine the molecular mechanism underlying this phenomenon.
Collapse
Affiliation(s)
- Evgeny Shmelkov
- a Department of Biochemistry and Molecular Pharmacology ; New York University School of Medicine ; New York , NY USA
| | | | | |
Collapse
|
87
|
Jang YH, Seong BL. Options and obstacles for designing a universal influenza vaccine. Viruses 2014; 6:3159-80. [PMID: 25196381 PMCID: PMC4147691 DOI: 10.3390/v6083159] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/13/2022] Open
Abstract
Since the discovery of antibodies specific to a highly conserved stalk region of the influenza virus hemagglutinin (HA), eliciting such antibodies has been considered the key to developing a universal influenza vaccine that confers broad-spectrum protection against various influenza subtypes. To achieve this goal, a prime/boost immunization strategy has been heralded to redirect host immune responses from the variable globular head domain to the conserved stalk domain of HA. While this approach has been successful in eliciting cross-reactive antibodies against the HA stalk domain, protective efficacy remains relatively poor due to the low immunogenicity of the domain, and the cross-reactivity was only within the same group, rather than among different groups. Additionally, concerns are raised on the possibility of vaccine-associated enhancement of viral infection and whether multiple boost immunization protocols would be considered practical from a clinical standpoint. Live attenuated vaccine hitherto remains unexplored, but is expected to serve as an alternative approach, considering its superior cross-reactivity. This review summarizes recent advancements in the HA stalk-based universal influenza vaccines, discusses the pros and cons of these approaches with respect to the potentially beneficial and harmful effects of neutralizing and non-neutralizing antibodies, and suggests future guidelines towards the design of a truly protective universal influenza vaccine.
Collapse
Affiliation(s)
- Yo Han Jang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, South Korea.
| | - Baik Lin Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, South Korea.
| |
Collapse
|
88
|
Pedersen NC. An update on feline infectious peritonitis: virology and immunopathogenesis. Vet J 2014; 201:123-32. [PMID: 24837550 PMCID: PMC7110662 DOI: 10.1016/j.tvjl.2014.04.017] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 03/24/2014] [Accepted: 04/27/2014] [Indexed: 11/18/2022]
Abstract
Feline infectious peritonitis (FIP) continues to be one of the most researched infectious diseases of cats. The relatively high mortality of FIP, especially for younger cats from catteries and shelters, should be reason enough to stimulate such intense interest. However, it is the complexity of the disease and the grudging manner in which it yields its secrets that most fascinate researchers. Feline leukemia virus infection was conquered in less than two decades and the mysteries of feline immunodeficiency virus were largely unraveled in several years. After a half century, FIP remains one of the last important infections of cats for which we have no single diagnostic test, no vaccine and no definitive explanations for how virus and host interact to cause disease. How can a ubiquitous and largely non-pathogenic enteric coronavirus transform into a highly lethal pathogen? What are the interactions between host and virus that determine both disease form (wet or dry) and outcome (death or resistance)? Why is it so difficult, and perhaps impossible, to develop a vaccine for FIP? What role do genetics play in disease susceptibility? This review will explore research conducted over the last 5 years that attempts to answer these and other questions. Although much has been learned about FIP in the last 5 years, the ultimate answers remain for yet more studies.
Collapse
Affiliation(s)
- Niels C Pedersen
- Center for Companion Animal Health, School of Veterinary Medicine, University of California, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
89
|
Evaluation of single-round infectious, chimeric dengue type 1 virus as an antigen for dengue functional antibody assays. Vaccine 2014; 32:4289-95. [DOI: 10.1016/j.vaccine.2014.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/19/2014] [Accepted: 06/06/2014] [Indexed: 12/26/2022]
|
90
|
Baykov IK, Matveev AL, Stronin OV, Ryzhikov AB, Matveev LE, Kasakin MF, Richter VA, Tikunova NV. A protective chimeric antibody to tick-borne encephalitis virus. Vaccine 2014; 32:3589-94. [DOI: 10.1016/j.vaccine.2014.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/28/2014] [Accepted: 05/01/2014] [Indexed: 12/30/2022]
|
91
|
Yip MS, Leung NHL, Cheung CY, Li PH, Lee HHY, Daëron M, Peiris JSM, Bruzzone R, Jaume M. Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus. Virol J 2014; 11:82. [PMID: 24885320 PMCID: PMC4018502 DOI: 10.1186/1743-422x-11-82] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 04/22/2014] [Indexed: 12/24/2022] Open
Abstract
Background Public health risks associated to infection by human coronaviruses remain considerable and vaccination is a key option for preventing the resurgence of severe acute respiratory syndrome coronavirus (SARS-CoV). We have previously reported that antibodies elicited by a SARS-CoV vaccine candidate based on recombinant, full-length SARS-CoV Spike-protein trimers, trigger infection of immune cell lines. These observations prompted us to investigate the molecular mechanisms and responses to antibody-mediated infection in human macrophages. Methods We have used primary human immune cells to evaluate their susceptibility to infection by SARS-CoV in the presence of anti-Spike antibodies. Fluorescence microscopy and real-time quantitative reverse transcriptase polymerase chain reaction (RT-PCR) were utilized to assess occurrence and consequences of infection. To gain insight into the underlying molecular mechanism, we performed mutational analysis with a series of truncated and chimeric constructs of fragment crystallizable γ receptors (FcγR), which bind antibody-coated pathogens. Results We show here that anti-Spike immune serum increased infection of human monocyte-derived macrophages by replication-competent SARS-CoV as well as Spike-pseudotyped lentiviral particles (SARS-CoVpp). Macrophages infected with SARS-CoV, however, did not support productive replication of the virus. Purified anti-viral IgGs, but not other soluble factor(s) from heat-inactivated mouse immune serum, were sufficient to enhance infection. Antibody-mediated infection was dependent on signaling-competent members of the human FcγRII family, which were shown to confer susceptibility to otherwise naïve ST486 cells, as binding of immune complexes to cell surface FcγRII was necessary but not sufficient to trigger antibody-dependent enhancement (ADE) of infection. Furthermore, only FcγRII with intact cytoplasmic signaling domains were competent to sustain ADE of SARS-CoVpp infection, thus providing additional information on the role of downstream signaling by FcγRII. Conclusions These results demonstrate that human macrophages can be infected by SARS-CoV as a result of IgG-mediated ADE and indicate that this infection route requires signaling pathways activated downstream of binding to FcγRII receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roberto Bruzzone
- HKU-Pasteur Research Pole and Center of Influenza Research, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR.
| | | |
Collapse
|
92
|
He J, Lai H, Engle M, Gorlatov S, Gruber C, Steinkellner H, Diamond MS, Chen Q. Generation and analysis of novel plant-derived antibody-based therapeutic molecules against West Nile virus. PLoS One 2014; 9:e93541. [PMID: 24675995 PMCID: PMC3968140 DOI: 10.1371/journal.pone.0093541] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/04/2014] [Indexed: 12/21/2022] Open
Abstract
Previously, our group engineered a plant-derived monoclonal antibody (MAb) (pHu-E16) that efficiently treated West Nile virus (WNV) infection in mice. In this study, we developed several pHu-E16 variants to improve its efficacy. These variants included a single-chain variable fragment (scFv) of pHu-E16 fused to the heavy chain (HC) constant domains (CH(1-3)) of human IgG (pHu-E16scFv-CH(1-3)) and a tetravalent molecule (Tetra pHu-E16) assembled from pHu-E16scFv-CH(1-3) with a second pHu-E16scFv fused to the light chain (LC) constant region. pHu-E16scFv-CH(1-3) and Tetra pHu-E16 were efficiently expressed and assembled in plants. To assess the impact of differences in N-linked glycosylation on pHu-E16 variant assembly and function, we expressed additional pHu-E16 variants with various combinations of HC and LC components. Our study revealed that proper pairing of HC and LC was essential for the complete N-glycan processing of antibodies in both plant and animal cells. Associated with their distinct N-glycoforms, pHu-E16, pHu-E16scFv-CH(1-3) and Tetra pHu-E16 exhibited differential binding to C1q and specific Fcγ receptors (FcγR). Notably, none of the plant-derived Hu-E16 variants showed antibody-dependent enhancement (ADE) activity in CD32A+ human cells, suggesting the potential of plant-produced antibodies to minimize the adverse effect of ADE. Importantly, all plant-derived MAb variants exhibited at least equivalent in vitro neutralization and in vivo protection in mice compared to mammalian cell-produced Hu-E16. This study demonstrates the capacity of plants to express and assemble a large, complex and functional IgG-like tetravalent mAb variant and also provides insight into the relationship between MAb N-glycosylation, FcγR and C1q binding, and ADE. These new insights may allow the development of safer and cost effective MAb-based therapeutics for flaviviruses, and possibly other pathogens.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/isolation & purification
- Complement C1q/immunology
- Complement C1q/metabolism
- Glycosylation
- Immunization, Passive
- Immunoconjugates/chemistry
- Immunoconjugates/genetics
- Immunoglobulin G/chemistry
- Immunoglobulin G/genetics
- Mice
- Mice, Inbred C57BL
- Plants, Genetically Modified
- Protein Binding
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Single-Chain Antibodies/administration & dosage
- Single-Chain Antibodies/biosynthesis
- Single-Chain Antibodies/isolation & purification
- Survival Analysis
- Nicotiana/genetics
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- West Nile Fever/immunology
- West Nile Fever/mortality
- West Nile Fever/prevention & control
- West Nile Fever/virology
- West Nile virus/immunology
- West Nile virus/pathogenicity
Collapse
Affiliation(s)
- Junyun He
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Huafang Lai
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Michael Engle
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Sergey Gorlatov
- MacroGenics, Inc, Rockville, Maryland, United States of America
| | - Clemens Gruber
- Department of Chemistry, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Herta Steinkellner
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Qiang Chen
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- College of Technology and Innovation, Arizona State University, Mesa, Arizona, United States of America
| |
Collapse
|
93
|
Cai Y, Ku Z, Liu Q, Leng Q, Huang Z. A combination vaccine comprising of inactivated enterovirus 71 and coxsackievirus A16 elicits balanced protective immunity against both viruses. Vaccine 2014; 32:2406-12. [PMID: 24657161 DOI: 10.1016/j.vaccine.2014.03.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/25/2014] [Accepted: 03/07/2014] [Indexed: 11/18/2022]
Abstract
Enterovirus 71 (EV71) and coxsackievirus A16 (CA16) are the two major causative agents of hand, foot and mouth disease (HFMD), which is an infectious disease frequently occurring in children. A bivalent vaccine against both EV71 and CA16 is highly desirable. In the present study, we compare monovalent inactivated EV71, monovalent inactivated CA16, and a combination vaccine candidate comprising of both inactivated EV71 and CA16, for their immunogenicity and in vivo protective efficacy. The two monovalent vaccines were found to elicit serum antibodies that potently neutralized the homologous virus but had no or weak neutralization activity against the heterologous one; in contrast, the bivalent vaccine immunized sera efficiently neutralized both EV71 and CA16. More importantly, passive immunization with the bivalent vaccine protected mice against either EV71 or CA16 lethal infections, whereas the monovalent vaccines only prevented the homologous but not the heterologous challenges. Together, our results demonstrate that the experimental bivalent vaccine comprising of inactivated EV71 and CA16 induces a balanced protective immunity against both EV71 and CA16, and thus provide proof-of-concept for further development of multivalent vaccines for broad protection against HFMD.
Collapse
Affiliation(s)
- Yicun Cai
- Center for Vaccine Sciences, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhiqiang Ku
- Center for Vaccine Sciences, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qingwei Liu
- Center for Vaccine Sciences, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qibin Leng
- Center for Vaccine Sciences, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhong Huang
- Center for Vaccine Sciences, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
94
|
Cook R, Leroux C, Issel C. Equine infectious anemia and equine infectious anemia virus in 2013: A review. Vet Microbiol 2013; 167:181-204. [DOI: 10.1016/j.vetmic.2013.09.031] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 09/16/2013] [Accepted: 09/21/2013] [Indexed: 10/26/2022]
|
95
|
Sjatha F, Takizawa Y, Kotaki T, Yamanaka A, Konishi E. Comparison of infection-neutralizing and -enhancing antibody balance induced by two distinct genotype strains of dengue virus type 1 or 3 DNA vaccines in mice. Microbes Infect 2013; 15:828-36. [DOI: 10.1016/j.micinf.2013.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 06/14/2013] [Accepted: 07/24/2013] [Indexed: 12/30/2022]
|
96
|
Tang D, Liu J, Li C, Zhang H, Ma P, Luo X, Zeng Z, Hong N, Liu X, Wang B, Wang F, Gan Z, Hao F. Positive effects of porcine IL-2 and IL-4 on virus-specific immune responses induced by the porcine reproductive and respiratory syndrome virus (PRRSV) ORF5 DNA vaccine in swine. J Vet Sci 2013; 15:99-109. [PMID: 24136204 PMCID: PMC3973771 DOI: 10.4142/jvs.2014.15.1.99] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 07/09/2013] [Indexed: 01/24/2023] Open
Abstract
The purpose of this study was to investigate the effects of porcine interleukin (IL)-2 and IL-4 genes on enhancing the immunogenicity of a porcine reproductive and respiratory syndrome virus ORF5 DNA vaccine in piglets. Eukaryotic expression plasmids pcDNA-ORF5, pcDNA-IL-2, and pcDNA-IL-4 were constructed and then expressed in Marc-145 cells. The effects of these genes were detected using an indirect immunofluorescent assay and reverse transcription polymerase chain reaction (RT-PCR). Characteristic fluorescence was observed at different times after pcDNA-ORF5 was expressed in the Marc-145 cells, and PCR products corresponding to ORF5, IL-2, and IL-4 genes were detected at 48 h. Based on these data, healthy piglets were injected intramuscularly with different combinations of the purified plasmids: pcDNA-ORF5 alone, pcDNA-ORF5 + pcDNA-IL-2, pcDNA-ORF5 + pcDNA-IL-4, and pcDNA-ORF5 + pcDNAIL-4 + pcDNA-IL-2. The ensuing humoral immune responses, percentages of CD4+ and CD8+ T lymphocytes, proliferation indices, and interferon-γ expression were analyzed. Results revealed that the piglets co-immunized with pcDNA-ORF5 + pcDNA-IL-4 + pcDNA-IL-2 plasmids developed significantly higher antibody titers and neutralizing antibody levels, had significantly increased levels of specific T lymphocyte proliferation, elevated percentages of CD4+ and CD8+ T lymphocytes, and significantly higher IFN-γ production than the other inoculated pigs (p < 0.05).
Collapse
Affiliation(s)
- Deyuan Tang
- Department of Animal Science, Guizhou University, Guiyang 550025,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
A mouse monoclonal antibody against dengue virus type 1 Mochizuki strain targeting envelope protein domain II and displaying strongly neutralizing but not enhancing activity. J Virol 2013; 87:12828-37. [PMID: 24049185 DOI: 10.1128/jvi.01874-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dengue fever and its more severe form, dengue hemorrhagic fever, are major global concerns. Infection-enhancing antibodies are major factors hypothetically contributing to increased disease severity. In this study, we generated 26 monoclonal antibodies (MAbs) against the dengue virus type 1 Mochizuki strain. We selected this strain because a relatively large number of unique and rare amino acids were found on its envelope protein. Although most MAbs showing neutralizing activities exhibited enhancing activities at subneutralizing doses, one MAb (D1-IV-7F4 [7F4]) displayed neutralizing activities without showing enhancing activities at lower concentrations. In contrast, another MAb (D1-V-3H12 [3H12]) exhibited only enhancing activities, which were suppressed by pretreatment of cells with anti-FcγRIIa. Although antibody engineering revealed that antibody subclass significantly affected 7F4 (IgG3) and 3H12 (IgG1) activities, neutralizing/enhancing activities were also dependent on the epitope targeted by the antibody. 7F4 recognized an epitope on the envelope protein containing E118 (domain II) and had a neutralizing activity 10- to 1,000-fold stronger than the neutralizing activity of previously reported human or humanized neutralizing MAbs targeting domain I and/or domain II. An epitope-blocking enzyme-linked immunosorbent assay (ELISA) indicated that a dengue virus-immune population possessed antibodies sharing an epitope with 7F4. Our results demonstrating induction of these antibody species (7F4 and 3H12) in Mochizuki-immunized mice may have implications for dengue vaccine strategies designed to minimize induction of enhancing antibodies in vaccinated humans.
Collapse
|
98
|
Simulations to compare efficacies of tetravalent dengue vaccines and mosquito vector control. Epidemiol Infect 2013; 142:1245-58. [PMID: 23925059 PMCID: PMC4045171 DOI: 10.1017/s0950268813001866] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SUMMARY Infection with dengue, the most prevalent mosquito-borne virus, manifests as dengue fever (DF) or the more fatal dengue haemorrhagic fever (DHF). DHF occurs mainly when an individual who has acquired antibodies to one serotype is inoculated with another serotype. It was reported that mosquito control may have increased the incidence of DF and DHF due to age-dependency in manifesting these illnesses or an immunological mechanism. Tetravalent dengue vaccine is currently being tested in clinical trials. However, seroconversions to all four serotypes were achieved only after three doses. Therefore, vaccines may predispose vaccinees to the risk of developing DHF in future infections. This study employed an individual-based computer simulation, to emulate mosquito control and vaccination, incorporating seroconversion rates reported from actual clinical trials. It was found that mosquito control alone would have increased incidence of DF and DHF in areas of high mosquito density. A vaccination programme with very high coverage, even with a vaccine of suboptimal seroconversion rates, attenuated possible surges in the incidence of DF and DHF which would have been caused by insufficient reduction in mosquito abundance. DHF cases attributable to vaccine-derived enhancement were fewer than DHF cases prevented by a vaccine with considerably high (although not perfect) seroconversion rates. These predictions may justify vaccination programmes, at least in areas of high mosquito abundance. In such areas, mosquito control programmes should be conducted only after the vaccination programme with a high coverage has been initiated.
Collapse
|
99
|
Rimmelzwaan GF, Fouchier RAM, Osterhaus ADME. Age distribution of cases caused by different influenza viruses. THE LANCET. INFECTIOUS DISEASES 2013; 13:646-7. [PMID: 23886322 PMCID: PMC7128308 DOI: 10.1016/s1473-3099(13)70181-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Guus F Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | |
Collapse
|
100
|
Li XQ, Qiu LW, Chen Y, Wen K, Cai JP, Chen J, Pan YX, Li J, Hu DM, Huang YF, Liu LD, Ding XX, Guo YH, Che XY. Dengue virus envelope domain III immunization elicits predominantly cross-reactive, poorly neutralizing antibodies localized to the AB loop: implications for dengue vaccine design. J Gen Virol 2013; 94:2191-2201. [PMID: 23851440 DOI: 10.1099/vir.0.055178-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-borne virus that causes severe health problems. An effective tetravalent dengue vaccine candidate that can provide life-long protection simultaneously against all four DENV serotypes is highly anticipated. A better understanding of the antibody response to DENV envelope protein domain III (EDIII) may offer insights into vaccine development. Here, we identified 25 DENV cross-reactive mAbs from immunization with Pichia pastoris-expressed EDIII of a single or all four serotype(s) using a prime-boost protocol, and through pepscan analysis found that 60 % of them (15/25) specifically recognized the same highly conserved linear epitope aa 309-320 of EDIII. All 15 complex-reactive mAbs exhibited significant cross-reactivity with recombinant EDIII from all DENV serotypes and also with C6/36 cells infected with DENV-1, -2, -3 and -4. However, neutralization assays indicated that the majority of these 15 mAbs were either moderately or weakly neutralizing. Through further epitope mapping by yeast surface display, two residues in the AB loop, Q316 and H317, were discovered to be critical. Three-dimensional modelling analysis suggests that this epitope is surface exposed on EDIII but less accessible on the surface of the E protein dimer and trimer, especially on the surface of the mature virion. It is concluded that EDIII as an immunogen may elicit cross-reactive mAbs toward an epitope that is not exposed on the virion surface, therefore contributing inefficiently to the mAbs neutralization potency. Therefore, the prime-boost strategy of EDIII from a single serotype or four serotypes mainly elicited a poorly neutralizing, cross-reactive antibody response to the conserved AB loop of EDIII.
Collapse
Affiliation(s)
- Xiao-Quan Li
- Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, PR China
| | - Li-Wen Qiu
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Yue Chen
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Kun Wen
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Jian-Piao Cai
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Jing Chen
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Yu-Xian Pan
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Jie Li
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Dong-Mei Hu
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Yan-Fen Huang
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Li-Dong Liu
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Xi-Xia Ding
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Yong-Hui Guo
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Xiao-Yan Che
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.,Laboratory of Emerging Infectious Diseases, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| |
Collapse
|