1
|
Wu Q, Chen K, Xue W, Wang G, Yang Y, Li S, Xia N, Chen Y. An insect cell-derived extracellular vesicle-based gB vaccine elicits robust adaptive immune responses against Epstein-Barr virus. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-023-2599-1. [PMID: 39499444 DOI: 10.1007/s11427-023-2599-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/19/2024] [Indexed: 11/07/2024]
Abstract
Epstein-Barr virus (EBV), the first identified human tumor virus, is implicated in various human malignancies, infectious mononucleosis, and more recently, multiple sclerosis. Prophylactic vaccines have the potential to effectively prevent EBV infection. Glycoprotein B (gB) serves as the fusogen and plays a pivotal role in the virus entry process, making it a critical target for EBV vaccine development. Surface membrane proteins of enveloped viruses serve as native conformational antigens, making them susceptible to immune recognition. Utilizing lipid membrane-bound viral antigens is a promising strategy for effective vaccine presentation in this context. In this study, we employed a truncated design for gB proteins, observing that these truncated gB proteins prompted a substantial release of extracellular vesicles (EVs) in insect cells. We verified that EVs exhibited abundant gB proteins, displaying the typical virus particle morphology and extracellular vesicle characteristics. gB EVs demonstrated a more efficient humoral and cellular immune response compared with the gB ectodomain trimer vaccine in mice. Moreover, the antisera induced by the gB EVs vaccine exhibited robust antibody-dependent cytotoxicity. Consequently, gB EVs-based vaccines hold significant potential for preventing EBV infection and offer valuable insights for vaccine design.
Collapse
Affiliation(s)
- Qian Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Kaiyun Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Wenhui Xue
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Guosong Wang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Yanbo Yang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Yixin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
2
|
Sehrawat S, Osterrieder N, Schmid DS, Rouse BT. Can the triumph of mRNA vaccines against COVID-19 be extended to other viral infections of humans and domesticated animals? Microbes Infect 2023; 25:105078. [PMID: 36435367 PMCID: PMC9682868 DOI: 10.1016/j.micinf.2022.105078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
The unprecedented success of mRNA vaccines in managing the COVID-19 pandemic raises the prospect of applying the mRNA platform to other viral diseases of humans and domesticated animals, which may lead to more efficacious vaccines for some agents. We briefly discuss reasons why mRNA vaccines achieved such success against COVID-19 and indicate what other virus infections and disease conditions might also be ripe for control using mRNA vaccines. We also evaluate situations where mRNA could prove valuable to rebalance the status of immune responsiveness and achieve success as a therapeutic vaccine approach against infections that induce immunoinflammatory lesions.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City, PO Manauli, Mohali 140306, Punjab, India.
| | - Nikolaus Osterrieder
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany; Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 5F, Block 1B, To Yuen Building, 31 To Yuen Street, Kowloon Tong, Hong Kong.
| | - D Scott Schmid
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee Knoxville, TN 37996-0845, USA.
| |
Collapse
|
3
|
Prevention of Congenital Cytomegalovirus Infection with Vaccines: State of the Art. Vaccines (Basel) 2021; 9:vaccines9050523. [PMID: 34069321 PMCID: PMC8158681 DOI: 10.3390/vaccines9050523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/15/2022] Open
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infection and non-genetic sensorineural hearing loss in childhood. Up to 2% of neonates, with the highest percentages found in developing countries, are congenitally infected with CMV. At birth, most of these infants are asymptomatic. However, approximately 10% have signs and symptoms of the disease, and 40–60% of symptomatic neonates will later develop permanent neurologic sequelae. To reduce congenital CMV (cCMV) infection, a vaccine able to prevent primary infection is essential. In this narrative review, actual ongoing research about the development of a CMV vaccine is discussed. The progressive increase in knowledge on the ways in which the host’s immune system and CMV relate has made it possible to clarify that the development of a vaccine that is certainly capable of reducing the risk of cCMV infection, and preventing both primary and nonprimary infections is extremely difficult. Many of the ways in which the virus evades the immune system and causes cCMV infection are not yet fully understood, especially in cases of nonprimary infection. Moreover, the schedule that should be recommended and that subjects must be vaccinated to obtain the greatest effect have not been precisely defined. Further studies are needed before the problem of cCMV infection and its related challenges can be totally solved.
Collapse
|
4
|
Xia W, Yan H, Zhang Y, Wang C, Gao W, Lv C, Wang W, Liu Z. Congenital Human Cytomegalovirus Infection Inducing Sensorineural Hearing Loss. Front Microbiol 2021; 12:649690. [PMID: 33936007 PMCID: PMC8079719 DOI: 10.3389/fmicb.2021.649690] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the primary cause of congenital infections. Despite its clinical significance, congenital HCMV infection is frequently overlooked clinically since most affected infants are asymptomatic. Sensorineural hearing loss (SNHL) is one of the most widely known disorders caused by congenital HCMV infection. The potential mechanism, however, remains unknown to date. The mechanism by which congenital HCMV infection induces sensorineural deafness has been partly characterized, leading to advancements in diagnosis, therapy, and prevention strategies. HCMV-induced hearing loss primarily involves immune responses, the release of inflammatory factors by natural killer (NK) cells, apoptosis of cochlear spiral ganglion, and potential changes due to vascular dysfunction. The diagnosis of HCMV induced SNHL includes serological examination to mothers, imaging, and amniotic fluid examination. Ganciclovir, mainly used for antiviral therapy and behavioral prevention, can, to some degree, prevent congenital HCMV infection. The role of HCMV infection in hearing loss needs further investigation since the mechanism of hearing loss caused by cytomegalovirus infection is not well understood. Although some advancement has been made in diagnosing and treating SNHL, more improvement is needed. A comprehensive understanding of cytomegalovirus’s pathogenesis is of key importance for preventing, diagnosing, and treating SNHL.
Collapse
Affiliation(s)
- Wenwen Xia
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Hui Yan
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yiyuan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Congcong Wang
- Department of Microbiology, Weifang Medical University, Weifang, China
| | - Wei Gao
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Changning Lv
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Wentao Wang
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zhijun Liu
- Department of Microbiology, Weifang Medical University, Weifang, China
| |
Collapse
|
5
|
Singh K, Hamilton ST, Shand AW, Hannan NJ, Rawlinson WD. Receptors in host pathogen interactions between human cytomegalovirus and the placenta during congenital infection. Rev Med Virol 2021; 31:e2233. [PMID: 33709529 DOI: 10.1002/rmv.2233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 11/09/2022]
Abstract
Cellular receptors in human cytomegalovirus (HCMV) mother to child transmission play an important role in congenital infection. Placental trophoblast cells are a significant cell type in placental development, placental functional processes, and in HCMV transmission. Different cells within the placental floating and chorionic villi present alternate receptors for HCMV cell entry. Syncytiotrophoblasts present neonatal Fc receptors that bind and transport circulating maternal immunoglobulin G across the placental interface which can also be bound to HCMV virions, facilitating viral entry into the placenta and foetal circulation. Cytotrophoblast express HCMV receptors including integrin-α1β1, integrin-αVβ3, epidermal growth factor receptor and platelet-derived growth factor receptor alpha. The latter interacts with HCMV glycoprotein-H, glycoprotein-L and glycoprotein-O (gH/gL/gO) trimers (predominantly in placental fibroblasts) and the gH/gL/pUL128, UL130-UL131A pentameric complex in other placental cell types. The pentameric complex allows viral tropism of placental trophoblasts, endothelial cells, epithelial cells, leukocytes and monocytes. This review outlines HCMV ligands and target receptor proteins in congenital HCMV infection.
Collapse
Affiliation(s)
- Krishneel Singh
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Stuart T Hamilton
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Antonia W Shand
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women Heidelberg, Victoria, Australia
| | - William D Rawlinson
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Singh T, Otero CE, Li K, Valencia SM, Nelson AN, Permar SR. Vaccines for Perinatal and Congenital Infections-How Close Are We? Front Pediatr 2020; 8:569. [PMID: 33384972 PMCID: PMC7769834 DOI: 10.3389/fped.2020.00569] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Congenital and perinatal infections are transmitted from mother to infant during pregnancy across the placenta or during delivery. These infections not only cause pregnancy complications and still birth, but also result in an array of pediatric morbidities caused by physical deformities, neurodevelopmental delays, and impaired vision, mobility and hearing. Due to the burden of these conditions, congenital and perinatal infections may result in lifelong disability and profoundly impact an individual's ability to live to their fullest capacity. While there are vaccines to prevent congenital and perinatal rubella, varicella, and hepatitis B infections, many more are currently in development at various stages of progress. The spectrum of our efforts to understand and address these infections includes observational studies of natural history of disease, epidemiological evaluation of risk factors, immunogen design, preclinical research of protective immunity in animal models, and evaluation of promising candidates in vaccine trials. In this review we summarize this progress in vaccine development research for Cytomegalovirus, Group B Streptococcus, Herpes simplex virus, Human Immunodeficiency Virus, Toxoplasma, Syphilis, and Zika virus congenital and perinatal infections. We then synthesize this evidence to examine how close we are to developing a vaccine for these infections, and highlight areas where research is still needed.
Collapse
Affiliation(s)
- Tulika Singh
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Claire E. Otero
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Katherine Li
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Sarah M. Valencia
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Ashley N. Nelson
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Sallie R. Permar
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
7
|
Guo B, Xu P, Chai D, Cao L, Liu L, Song T, Hu S, Chen Y, Yan X, Xu T. gB co-immunization with GP96 enhances pulmonary-resident CD8 T cells and exerts a long-term defence against MCMV pneumonitis. J Cell Mol Med 2020; 24:14426-14440. [PMID: 33155438 PMCID: PMC7754068 DOI: 10.1111/jcmm.16065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection in the respiratory tract leads to pneumonitis in immunocompromised hosts without available vaccine. Considering cytomegalovirus (CMV) mainly invades through the respiratory tract, CMV-specific pulmonary mucosal vaccine development that provides a long-lasting protection against CMV challenge gains our attention. In this study, N-terminal domain of GP96 (GP96-NT) was used as a mucosal adjuvant to enhance the induction of pulmonary-resident CD8 T cells elicited by MCMV glycoprotein B (gB) vaccine. Mice were intranasally co-immunized with 50 μg pgB and equal amount of pGP96-NT vaccine 4 times at 2-week intervals, and then i.n. challenged with MCMV at 16 weeks after the last immunization. Compared with pgB immunization alone, co-immunization with pgB/pGP96-NT enhanced a long-lasting protection against MCMV pneumonitis by significantly improved pneumonitis pathology, enhanced bodyweight, reduced viral burdens and increased survival rate. Moreover, the increased CD8 T cells were observed in lung but not spleen from pgB/pGP96-NT co-immunized mice. The increments of pulmonary CD8 T cells might be mainly due to non-circulating pulmonary-resident CD8 T-cell subset expansion but not circulating CD8 T-cell populations that home to inflammation site upon MCMV challenge. Finally, the deterioration of MCMV pneumonitis by depletion of pulmonary site-specific CD8 T cells in mice that were pgB/pGP96-NT co-immunization might be a clue to interpret the non-circulating pulmonary-resident CD8 T subset expansion. These data might uncover a promising long-lasting prophylactic vaccine strategy against MCMV-induced pneumonitis.
Collapse
Affiliation(s)
- Bingnan Guo
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Peifeng Xu
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Lei Cao
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lin Liu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tengfei Song
- The Feinstein Institute for Medical Research, Manhasset, New York, NY, USA
| | - Shuqun Hu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuling Chen
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xianliang Yan
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tie Xu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China.,Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Emergency, Nanjing Jiangning Hospital, Nanjing, China
| |
Collapse
|
8
|
Shibamura M, Yoshikawa T, Yamada S, Inagaki T, Nguyen PHA, Fujii H, Harada S, Fukushi S, Oka A, Mizuguchi M, Saijo M. Association of human cytomegalovirus (HCMV) neutralizing antibodies with antibodies to the HCMV glycoprotein complexes. Virol J 2020; 17:120. [PMID: 32746933 PMCID: PMC7397426 DOI: 10.1186/s12985-020-01390-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) causes asymptomatic infections, but also causes congenital infections when women were infected with HCMV during pregnancy, and life-threatening diseases in immunocompromised patients. To better understand the mechanism of the neutralization activity against HCMV, the association of HCMV NT antibody titers was assessed with the antibody titers against each glycoprotein complex (gc) of HCMV. Methods Sera collected from 78 healthy adult volunteers were used. HCMV Merlin strain and HCMV clinical isolate strain 1612 were used in the NT assay with the plaque reduction assay, in which both the MRC-5 fibroblasts cells and the RPE-1 epithelial cells were used. Glycoprotein complex of gB, gH/gL complexes (gH/gL/gO and gH/gL/UL128–131A [PC]) and gM/gN were selected as target glycoproteins. 293FT cells expressed with gB, gM/gN, gH/gL/gO, or PC, were prepared and used for the measurement of the antibody titers against each gc in an indirect immunofluorescence assay (IIFA). The correlation between the IIFA titers to each gc and the HCMV-NT titers was evaluated. Results There were no significant correlations between gB-specific IIFA titers and the HCMV-NT titers in epithelial cells or between gM/gN complex-specific IIFA titers and the HCMV-NT titers. On the other hand, there was a statistically significant positive correlation between the IIFA titers to gH/gL complexes and HCMV-NT titers. Conclusions The data suggest that the gH/gL complexes might be the major target to induce NT activity against HCMV.
Collapse
Affiliation(s)
- Miho Shibamura
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.,Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Souichi Yamada
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Takuya Inagaki
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.,Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, Japan
| | - Phu Hoang Anh Nguyen
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.,Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Hikaru Fujii
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.,The Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
| | - Shizuko Harada
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Shuetsu Fukushi
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akira Oka
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masashi Mizuguchi
- Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan. .,Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
9
|
Cui X, Cao Z, Wang S, Adler SP, McVoy MA, Snapper CM. Immunization with Human Cytomegalovirus Core Fusion Machinery and Accessory Envelope Proteins Elicit Strong Synergistic Neutralizing Activities. Vaccines (Basel) 2020; 8:vaccines8020179. [PMID: 32294946 PMCID: PMC7348949 DOI: 10.3390/vaccines8020179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/04/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) core fusion machinery proteins gB and gH/gL, and accessory proteins UL128/UL130/UL131A, are the key envelope proteins that mediate HCMV entry into and infection of host cells. To determine whether these HCMV envelope proteins could elicit neutralizing activities synergistically, we immunized rabbits with individual or various combinations of these proteins adsorbed to aluminum hydroxide mixed with CpG-ODN. We then analyzed serum neutralizing activities with multiple HCMV laboratory strains and clinical isolates. HCMV trimeric gB and gH/gL elicited high and moderate titers of HCMV neutralizing activity, respectively. HCMV gB in combination with gH/gL elicited up to 17-fold higher HCMV neutralizing activities compared to the sum of neutralizing activity elicited by the individual proteins analyzed with both fibroblasts and epithelial cells. HCMV gB+gH/gL+UL128/UL130/UL131A in combination increased the neutralizing activity up to 32-fold compared to the sum of neutralizing activities elicited by the individual proteins analyzed with epithelial cells. Adding UL128/UL130/UL131A to gB and gH/gL combination did not increase further the HCMV neutralizing activity analyzed with fibroblasts. These data suggest that the combination of HCMV core fusion machinery envelope proteins gB+gH/gL or the combination of gB and pentameric complex could be ideal vaccine candidates that would induce optimal immune responses against HCMV infection.
Collapse
Affiliation(s)
- Xinle Cui
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3498
| | - Zhouhong Cao
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shuishu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Michael A. McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Clifford M. Snapper
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
10
|
Nganga SW, Otieno NA, Adero M, Ouma D, Chaves SS, Verani JR, Widdowson MA, Wilson A, Bergenfeld I, Andrews C, Fenimore VL, Gonzalez-Casanova I, Frew PM, Omer SB, Malik FA. Patient and provider perspectives on how trust influences maternal vaccine acceptance among pregnant women in Kenya. BMC Health Serv Res 2019; 19:747. [PMID: 31651307 PMCID: PMC6813986 DOI: 10.1186/s12913-019-4537-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/16/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Pregnant women and newborns are at high risk for infectious diseases. Altered immunity status during pregnancy and challenges fully vaccinating newborns contribute to this medical reality. Maternal immunization is a strategy to protect pregnant women and their newborns. This study aimed to find out how patient-provider relationships affect maternal vaccine uptake, particularly in the context of a lower middle- income country where limited research in this area exists. METHODS We conducted semi-structured, in-depth narrative interviews of both providers and pregnant women from four sites in Kenya: Siaya, Nairobi, Mombasa, and Marsabit. Interviews were conducted in either English or one of the local regional languages. RESULTS We found that patient trust in health care providers (HCPs) is integral to vaccine acceptance among pregnant women in Kenya. The HCP-patient relationship is a fiduciary one, whereby the patients' trusts is primarily rooted in the provider's social position as a person who is highly educated in matters of health. Furthermore, patient health education and provider attitudes are crucial for reinstating and fostering that trust, especially in cases where trust was impeded by rumors, community myths and misperceptions, and religious and cultural factors. CONCLUSION Patient trust in providers is a strong facilitator contributing to vaccine acceptance among pregnant women in Kenya. To maintain and increase immunization trust, providers have a critical role in cultivating a positive environment that allows for favorable interactions and patient health education. This includes educating providers on maternal immunizations and enhancing knowledge of effective risk communication tactics in clinical encounters.
Collapse
Affiliation(s)
- Stacy W Nganga
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Nancy A Otieno
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Maxwell Adero
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Dominic Ouma
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Sandra S Chaves
- Division of Global Health Protection, Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Jennifer R Verani
- Division of Global Health Protection, Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Marc-Alain Widdowson
- Division of Global Health Protection, Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Andrew Wilson
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Irina Bergenfeld
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Courtni Andrews
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Vincent L Fenimore
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA.,UNLV School of Public Health, University of Nevada, Las Vegas, NV, USA
| | - Ines Gonzalez-Casanova
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA.
| | - Paula M Frew
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA.,UNLV School of Public Health, University of Nevada, Las Vegas, NV, USA.,Department of Behavioral Science and Health Education, Emory University Rollins School of Public Health, Atlanta, GA, USA.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.,UNLV Population Health & Health Equity Initiative, University of Nevada, Las Vegas, NV, USA
| | - Saad B Omer
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA.,Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA.,Division of Pediatrics, Atlanta, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Fauzia A Malik
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| |
Collapse
|
11
|
Saadatnia G, Saremi S, Salehian B, Salehian P. Uterine Leiomyoma and Reproductive Tract Infections Detected by Polymerase Chain Reaction. IRANIAN JOURNAL OF PATHOLOGY 2019; 14:33-40. [PMID: 31531099 PMCID: PMC6708570 DOI: 10.30699/ijp.14.1.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/24/2018] [Indexed: 11/16/2022]
Abstract
Background and Objective: For nearly a century, it has been suspected that reproductive tract infections play an etio- logic role in uterine leiomyoma. However, no epidemiologic study of leiomyoma has used the polymerase chain reaction (PCR) to compare uterine tissues from cases and non-cases, and to investigate associations between uterine leiomyoma and infections detected by PCR. Methods: In this case-control study, 92 leiomyoma tissues from cases, and 94 myometrial tissue from controls were screened by PCR for cytomegalovirus, Chlamydia trachomatis, herpes simplex virus-1, 2, and human papillomavirus typed as 16/18 or another strain. Multivariable analysis used age-adjusted logistic regression, and generalized linear regression as appropriate. Results: In the uterine tissues of cases and unmatched controls, the prevalence of infection was: cytomegalovirus (32.6%, 7.4%), C. trachomatis (23.9%, 37.2%), herpes simplex virus-1,2 (25.0%, 13.8%), human papillomavirus 16/18 (13.0%, 10.5%). Leiomyoma was associated with cytomegalovirus (Odds Ratio (O.R.) 6.10; 95% confidence interval (C.I.), 2.40, 15.55) and Chlamydia (O.R. 0.47; 95% C.I. 0.23, 0.97). Likewise, the log count of leiomyoma was higher with cytomegalovirus (+0.65, 95% C.I. +0.34, +0.95) and lower with Chlamydia (-0.71, 95% C.I. -1.12, -0.29). Conclusion: This first application of PCR to leiomyomata and control uterine tissues from non-cases reveals that cytomegalovirus is associated with the presence, number, and volume of uterine leiomyoma, while C. trachomatis is inversely associated with leiomyoma, but only in the absence of cytomegalovirus. Current findings provide preliminary evidence that common reproductive tract infections contribute to the growth and control of at least some cases of uterine leiomyoma.
Collapse
Affiliation(s)
- Geita Saadatnia
- Dept. of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Saadatnia Saremi
- Sarem Cell Research Center, Sarem Women's Hospital, Tehran, Iran
| | - Behrouz Salehian
- Dept. of Endocrinology, City of Hope National Medical Center, Duarte, California
| | | |
Collapse
|
12
|
Cui X, Cao Z, Wang S, Flora M, Adler SP, McVoy MA, Snapper CM. Immunization of Rabbits with Recombinant Human Cytomegalovirus Trimeric versus Monomeric gH/gL Protein Elicits Markedly Higher Titers of Antibody and Neutralization Activity. Int J Mol Sci 2019; 20:ijms20133158. [PMID: 31261659 PMCID: PMC6651862 DOI: 10.3390/ijms20133158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 11/30/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection and HCMV infection of immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. HCMV envelope glycoproteins gB, gH, and gL, which constitute the core fusion machinery, play critical roles in HCMV fusion and entry into host cells. HCMV gB and gH/gL have been reported to elicit potent neutralizing antibodies. Recently, the gB/gH/gL complex was identified in the envelope of HCMV virions, and 16–50% of the total gH/gL bound to gB, forming the gB/gH/gL complex. These findings make the gB/gH/gL a unique HCMV vaccine candidate. We previously reported the production of HCMV trimeric gB and gH/gL heterodimers, and immunization with a combination of trimeric gB and gH/gL heterodimers elicited strong synergistic HCMV-neutralizing activity. To further improve the immunogenicity of gH/gL, we produced trimeric gH/gL. Rabbits immunized with HCMV trimeric gH/gL induced up to 38-fold higher serum titers of gH/gL-specific IgG relative to HCMV monomeric gH/gL, and elicited ~10-fold higher titers of complement-dependent and complement-independent HCMV-neutralizing activity for both epithelial cells and fibroblasts. HCMV trimeric gH/gL in combination with HCMV trimeric gB would be a novel promising HCMV vaccine candidate that could induce highly potent neutralizing activities.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Zhouhong Cao
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shuishu Wang
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Michael Flora
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
13
|
Abstract
Congenital human cytomegalovirus (HCMV) infection and HCMV infection of the immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. Efforts to develop HCMV vaccines have been ongoing for 50 y, though no HCMV vaccine has been licensed; encouraging and promising results have obtained from both preclinical and clinical trials. HCMV infection induces a wide range of humoral and T cell-mediated immune responses, and both branches of immunity are correlated with protection. In recent years, there have been novel approaches toward the development of HCMV vaccines and demonstrated that vaccine candidates could potentially provide superior protection over natural immunity acquired following HCMV infection. Further, rationally designed HCMV protein antigens that express native conformational epitopes could elicit optimal immune response. HCMV vaccine candidates, using a multi-antigen approach, to maximize the elicited protective immunity will most likely be successful in development of HCMV vaccine.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
14
|
[A review on the prevention and treatment of congenital cytomegalovirus infection in mothers and infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20. [PMID: 30369367 PMCID: PMC7389047 DOI: 10.7499/j.issn.1008-8830.2018.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Human cytomegalovirus (HCMV) has a high infection rate worldwide, and 85%-90% of congenital cytomegalovirus (CMV) infections are asymptomatic at birth, with the clinical manifestations of hearing loss, psychomotor retardation, and learning disabilities, while 10%-15% are symptomatic infections. Some preterm infants develop CMV infection after birth, which can cause sepsis-like syndrome, thrombocytopenia, neutropenia, liver injury, and lung injury. However at present, women of childbearing age have a lack of awareness of CMV. CMV education and hygiene precautions for pregnant women can prevent CMV infections in themselves and congenital CMV infections in their infants. No definite results have been obtained from the studies on the effect of CMV vaccine and high-titer immunoglobulin in preventing congenital CMV infection in fetuses. Recent studies have confirmed that the specificity and sensitivity of urinary or salivary CMV-DNA detection have reached more than 98%, which contributes to the early diagnosis of congenital CMV infection. In addition to short-term treatment with ganciclovir, long-term treatment with oral valganciclovir is safe for symptomatic congenital CMV infection and appears to have a better clinical effect than the short-term treatment. In the future, it is necessary to strengthen the health education for pregnant women, enhance the mother-to-child management of CMV infection, conduct the research on CMV vaccine, and further standardize treatment regimens.
Collapse
|
15
|
Cui X, Cao Z, Wang S, Lee RB, Wang X, Murata H, Adler SP, McVoy MA, Snapper CM. Novel trimeric human cytomegalovirus glycoprotein B elicits a high-titer neutralizing antibody response. Vaccine 2018; 36:5580-5590. [PMID: 30082162 PMCID: PMC6556890 DOI: 10.1016/j.vaccine.2018.07.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/03/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Human cytomegalovirus (HCMV) is a major cause of disability in congenitally infected infants and in the immunosuppressed. There is currently no licensed prophylactic HCMV vaccine. The HCMV envelope glycoprotein B (gB) is considered a major vaccine target antigen based on its critical role in mediating viral-host cell fusion and thus viral entry. The natural conformation of HCMV gB within the viral envelope is a trimer, but there has been no reported success in producing a recombinant trimeric gB suitable for vaccine use. Phase II clinical trials of a monomeric recombinant gB protein demonstrated 50% efficacy in preventing HCMV infection in seronegative women of reproductive age, and in reducing viremia in solid organ transplantation recipients. We now report the production of a uniformly trimeric recombinant HCMV gB protein in Chinese ovary cells, as demonstrated by Western blot analysis under modified non-reducing conditions and size exclusion chromatography with multi-angle scattering. Immunization of mice with trimeric HCMV gB induced up to 11-fold higher serum titers of total gB-specific IgG relative to monomeric HCMV gB using Alum + CpG as adjuvants. Further, trimeric HCMV gB elicited 50-fold higher complement-independent and 20-fold higher complement-dependent HCMV neutralizing titers compared to monomeric HCMV gB using the fibroblast cell line, MRC-5, and up to 6-fold higher complement-independent and -dependent HCMV neutralizing titers using the epithelial cell line, ARPE-19. The markedly enhanced HCMV neutralizing activity in response to trimeric HCMV gB was also observed using an additional four distinct clinical HCMV isolates. These data support a role for trimeric HCMV gB as an important component for clinical testing of a prophylactic HCMV vaccine.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States.
| | - Zhouhong Cao
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Shuishu Wang
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Ronzo B Lee
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Xiao Wang
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Haruhiko Murata
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Stuart P Adler
- CMV Research Foundation, Richmond VA 23229, United States
| | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| |
Collapse
|
16
|
Efficient Delivery of Human Cytomegalovirus T Cell Antigens by Attenuated Sendai Virus Vectors. J Virol 2018; 92:JVI.00569-18. [PMID: 29769344 DOI: 10.1128/jvi.00569-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) represents a major cause of clinical complications during pregnancy as well as immunosuppression, and the licensing of a protective HCMV vaccine remains an unmet global need. Here, we designed and validated novel Sendai virus (SeV) vectors delivering the T cell immunogens IE-1 and pp65. To enhance vector safety, we used a replication-deficient strain (rdSeV) that infects target cells in a nonproductive manner while retaining viral gene expression. In this study, we explored the impact that transduction with rdSeV has on human dendritic cells (DCs) by comparing it to the parental, replication-competent Sendai virus strain (rcSeV) as well as the poxvirus strain modified vaccinia Ankara (MVA). We found that wild-type SeV is capable of replicating to high titers in DCs while rdSeV infects cells abortively. Due to the higher degree of attenuation, IE-1 and pp65 protein levels mediated by rdSeV after infection of DCs were markedly reduced compared to those of the parental Sendai virus recombinants, but antigen-specific restimulation of T cell clones was not negatively affected by this. Importantly, rdSeV showed reduced cytotoxic effects compared to rcSeV and MVA and was capable of mediating DC maturation as well as secretion of alpha interferon and interleukin-6. Finally, in a challenge model with a murine cytomegalovirus (MCMV) strain carrying an HCMV pp65 peptide, we found that viral replication was restricted if mice were previously vaccinated with rdSeV-pp65. Taken together, these data demonstrate that rdSeV has great potential as a vector system for the delivery of HCMV immunogens.IMPORTANCE HCMV is a highly prevalent betaherpesvirus that establishes lifelong latency after primary infection. Congenital HCMV infection is the most common viral complication in newborns, causing a number of late sequelae ranging from impaired hearing to mental retardation. At the same time, managing HCMV reactivation during immunosuppression remains a major hurdle in posttransplant care. Since options for the treatment of HCMV infection are still limited, the development of a vaccine to confine HCMV-related morbidities is urgently needed. We generated new vaccine candidates in which the main targets of T cell immunity during natural HCMV infection, IE-1 and pp65, are delivered by a replication-deficient, Sendai virus-based vector system. In addition to classical prophylactic vaccine concepts, these vectors could also be used for therapeutic applications, thereby expanding preexisting immunity in high-risk groups such as transplant recipients or for immunotherapy of glioblastomas expressing HCMV antigens.
Collapse
|
17
|
Chandrasekaran AP, Song M, Kim KS, Ramakrishna S. Different Methods of Delivering CRISPR/Cas9 Into Cells. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 159:157-176. [PMID: 30340786 DOI: 10.1016/bs.pmbts.2018.05.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system (Cas) is comprised of repetitive bases followed by short fragments of DNA from a previously invading organism that provide immunity to the most prokaryotic organisms. An RNA-dependent spacer is required for CRISPR/Cas9 to recognize the target DNA. Delivery of the CRISPR/Cas9-guide RNA (gRNA) complex to any cell results in modification of the target sequence. The CRISPR/Cas9-mediated genome editing technique is currently in the spotlight and has several research interests, including molecular medicine and agriculture. There are several factors that hinder the delivery of this complex, such as the large size of the plasmid or high dosage of the chemical agent. There are several methods available to deliver CRISPR/Cas9 and its components to the target cells. It includes viral, non-viral and physical methods to deliver plasmid or ribonucleoprotein (RNP) of CRISPR components. But in vivo CRISPR/Cas9 delivery remains challenging to the researchers due to insertional mutagenesis, targeted delivery, immunogenicity, and off-targets. However, studies suggesting that the CRISPR/Cas9-RNP delivery can overcome these hurdles. Here, we review the various methods for delivery of CRISPR/Cas9 and gRNA to several cell lines, highlighting the limitations of each approach, and suggest possible alternative methods.
Collapse
Affiliation(s)
| | - Minjung Song
- Department of Food Biotechnology, College of Medical and Life Science, Silla University, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| |
Collapse
|
18
|
Viral infections in solid organ transplant recipients: novel updates and a review of the classics. Curr Opin Infect Dis 2018; 30:579-588. [PMID: 28984642 DOI: 10.1097/qco.0000000000000409] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW To summarize new discoveries in viral pathogenesis and novel therapeutic and prophylactic strategies in organ transplant recipients. RECENT FINDINGS For decades, prophylaxis of cytomegalovirus (CMV) has been the standard preventive strategy, but new clinical trials are expected to determine the advantages of preemptive therapy over prophylaxis. Novel anti-CMV agents, such as maribavir and letermovir, are being studied for the treatment of resistant/refractory CMV as alternatives to foscarnet and cidofovir. CMV immune monitoring may offer individualized management plans. Epstein-Barr virus infections in transplant recipients are difficult to prevent and treat, though recent data suggest possible merit to pretransplant rituximab among high-risk transplant recipients. We review the groundbreaking HIV-to-HIV organ transplant trials, which are expected to revolutionize the care of HIV-infected individuals. Finally, we review topical developments in human herpesvirus 8, Zika virus, RNA respiratory viruses, adenovirus, norovirus, and polyoma viruses in organ transplantation. SUMMARY Ongoing trials to optimize CMV prophylaxis and treatment, and outcomes of HIV-to-HIV organ transplantation in the United States, have significant implications to optimize management of these viruses in transplant recipients. Assessment of new antivirals and antiviral strategies, such as adoptive immunotherapy, is warranted for refractory viral infections.
Collapse
|
19
|
Kruger MM, Martin LJ, Maistry S, Heathfield LJ. A systematic review exploring the relationship between infection and sudden unexpected death between 2000 and 2016: A forensic perspective. Forensic Sci Int 2018; 289:108-119. [PMID: 29860163 DOI: 10.1016/j.forsciint.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 11/30/2022]
Abstract
Death due to infectious diseases is a major health concern worldwide. This is of particular concern in developing countries where poor-socio economic status and a lack of healthcare resources contribute to the high burden of disease. In some cases death due to infection can be acute and aggressive, and death may occur without a diagnosis whilst the person is still alive. These deaths may ultimately lead to a medico-legal autopsy being performed. There are various mechanisms by which sudden death due to infection may occur. In addition, there are many risk factors associated with sudden death due to infection, which differ between infants and older individuals. However, it is unclear which pathogens and risk factors are most frequently associated with sudden death due to infection. Therefore a systematic review of articles and case reports published between 1 January 2000 and 30 June 2016 was undertaken in order to (1) explore the relationship between pathogens and their causative role and (2) identify the relationship between predisposing and/or risk factors associated with sudden death due to infection. Major databases were searched and after critical appraisal 143 articles were identified. It was found that respiratory infections and deaths involving bacterial pathogens were most commonly associated with these deaths. In addition the most common risk factors in infants were exposure to tobacco smoke and co-sleeping. In adults the most common risk factors were co-morbid conditions and illnesses. This information aids in a better understanding of these deaths and highlights the need for more research in this field, particularly in developing countries.
Collapse
Affiliation(s)
- Mia M Kruger
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa.
| | - Lorna J Martin
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa.
| | - Sairita Maistry
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa.
| | - Laura J Heathfield
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa.
| |
Collapse
|
20
|
Asian Elephant T Cell Responses to Elephant Endotheliotropic Herpesvirus. J Virol 2018; 92:JVI.01951-17. [PMID: 29263271 PMCID: PMC5827410 DOI: 10.1128/jvi.01951-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
Elephant endotheliotropic herpesvirus (EEHV) can cause lethal hemorrhagic disease in juvenile Asian elephants, an endangered species. One hypothesis to explain this vulnerability of some juvenile elephants is that they fail to mount an effective T cell response to the virus. To our knowledge, there have been no studies of Asian elephant T cell responses to EEHV. To address this deficiency, we validated the gamma interferon (IFN-γ) enzyme-linked immunospot assay for tracking antigen-directed T cell activity by monitoring rabies-specific responses in vaccinated elephants. In addition, we generated monoclonal antibodies to Asian elephant CD4 and CD8 to facilitate phenotypic T cell profiling. Using these tools, we screened healthy elephants with a history of EEHV infection for reactivity against nine EEHV proteins whose counterparts in other herpesviruses are known to induce T cell responses in their natural hosts. We identified glycoprotein B (gB) and the putative regulatory protein E40 as the most immunogenic T cell targets (IFN-γ responses in five of seven elephants), followed by the major capsid protein (IFN-γ responses in three of seven elephants). We also observed that IFN-γ responses were largely from CD4+ T cells. We detected no activity against the predicted major immediate early (E44) and large tegument (E34) proteins, both immunodominant T cell targets in humans latently infected with cytomegalovirus. These studies identified EEHV-specific T cells in Asian elephants for the first time, lending insight into the T cell priming that might be required to protect against EEHV disease, and will guide the design of effective vaccine strategies. IMPORTANCE Endangered Asian elephants are facing many threats, including lethal hemorrhagic disease from elephant endotheliotropic herpesvirus (EEHV). EEHV usually establishes chronic, benign infections in mature Asian elephants but can be lethal to juvenile elephants in captivity and the wild. It is the leading cause of death in captive Asian elephants in North America and Europe. Despite the availability of sensitive tests and protocols for treating EEHV-associated illness, these measures are not always effective. The best line of defense would be a preventative vaccine. We interrogated normal healthy elephants previously infected with EEHV for T cell responses to nine EEHV proteins predicted to induce cellular immune responses. Three proteins elicited IFN-γ responses, suggesting their potential usefulness as vaccine candidates. Our work is the first to describe T cell responses to a member of the proposed fourth subfamily of mammalian herpesviruses, the Deltaherpesvirinae, within a host species in the clade Afrotheria. An EEHV vaccine would greatly contribute to the health care of Asian and African elephants that are also susceptible to this disease.
Collapse
|
21
|
Humanes Cytomegalievirus (HCMV). Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2018; 61:116-128. [DOI: 10.1007/s00103-017-2661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
22
|
Itell HL, Nelson CS, Martinez DR, Permar SR. Maternal immune correlates of protection against placental transmission of cytomegalovirus. Placenta 2017; 60 Suppl 1:S73-S79. [PMID: 28456432 PMCID: PMC5650553 DOI: 10.1016/j.placenta.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/07/2017] [Accepted: 04/13/2017] [Indexed: 02/02/2023]
Abstract
Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. In a subset of infected infants, congenital HCMV causes severe, long-lasting sequelae, including deafness, microcephaly, neurodevelopmental delay, and even death. Accordingly, a maternal vaccine to prevent congenital HCMV infection continues to be a top public health priority. Nevertheless, all vaccines tested to date have failed to meet clinical trial endpoints. Maternal immunity provides partial protection against congenital HCMV transmission, as vertical transmission from seropositive mothers is relatively rare. Therefore, an understanding of the maternal immune correlates of protection against HCMV congenital infection will be critical to inform design of an efficacious maternal vaccine. This review summarizes our understanding of the innate and adaptive immune correlates of protection against congenital transmission of HCMV, and discusses the advantages and applications of a novel nonhuman primate model of congenital CMV transmission to aid in rational vaccine design and evaluation.
Collapse
Affiliation(s)
- Hannah L Itell
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Cody S Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - David R Martinez
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
23
|
Abstract
Cytomegalovirus (CMV) is a frequent complication of hematopoietic stem cell transplant in pediatric patients, with significant morbidity and mortality. Antiviral drugs are used as prophylactic, preemptive or therapeutic medicines; however, no uniform guidelines exist for the best strategy to prevent CMV disease. Resistance to standard antiviral therapies can lead to further difficulty in managing CMV disease. Studies for investigational therapies are underway and could provide options for treatment of resistant CMV, while limiting toxicities associated with currently used antiviral therapies.
Collapse
|
24
|
Vohr BR, Poggi Davis E, Wanke CA, Krebs NF. Neurodevelopment: The Impact of Nutrition and Inflammation During Preconception and Pregnancy in Low-Resource Settings. Pediatrics 2017; 139:S38-S49. [PMID: 28562247 DOI: 10.1542/peds.2016-2828f] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2016] [Indexed: 11/24/2022] Open
Abstract
The rapid pace of fetal development by far exceeds any other stage of the life span, and thus, environmental influences can profoundly alter the developmental course. Stress during the prenatal period, including malnutrition and inflammation, impact maternal and fetal neurodevelopment with long-term consequences for physical and mental health of both the mother and her child. One primary consequence of maternal malnutrition, inflammation, and other sources of prenatal stress is a poor birth outcome, such as prematurity or growth restriction. These phenotypes are often used as indications of prenatal adversity. In fact, the original evidence supporting the fetal programming hypothesis came from studies documenting an association between birth phenotype and the development of subsequent physical and mental health problems. Fetal growth restriction in both term and preterm infants is associated with neonatal morbidities and a wide variety of behavioral and psychological diagnoses in childhood and adolescence, including attention-deficit/hyperactivity disorder, anxiety, depression, internalizing and thought problems, poor social skills, and autism spectrum disorder. Improving maternal-child health requires interventions that begin before pregnancy and continue throughout gestation and into the postpartum period. Such interventions might include supporting pregnancy intention, maternal nutrition, health/medical care, mental health, and providing social support. This article discusses the impact of maternal nutrition and inflammation during preconception and pregnancy among women living in low-resource settings, with an emphasis on key knowledge gaps that need to be addressed to guide program and policy decisions at local, regional and global levels.
Collapse
Affiliation(s)
- Betty R Vohr
- Neonatal Follow-up Program, Women & Infants Hospital, Providence, Rhode Island; .,Department of Pediatrics, Brown University, Providence, Rhode Island
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, Colorado.,Department of Psychiatry, University of California Irvine, Irvine, California
| | - Christine A Wanke
- Department of Public Health and Community Medicine, Tufts University, Medford, Massachusetts; and
| | - Nancy F Krebs
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
25
|
Jean Beltran PM, Federspiel JD, Sheng X, Cristea IM. Proteomics and integrative omic approaches for understanding host-pathogen interactions and infectious diseases. Mol Syst Biol 2017; 13:922. [PMID: 28348067 PMCID: PMC5371729 DOI: 10.15252/msb.20167062] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Organisms are constantly exposed to microbial pathogens in their environments. When a pathogen meets its host, a series of intricate intracellular interactions shape the outcome of the infection. The understanding of these host–pathogen interactions is crucial for the development of treatments and preventive measures against infectious diseases. Over the past decade, proteomic approaches have become prime contributors to the discovery and understanding of host–pathogen interactions that represent anti‐ and pro‐pathogenic cellular responses. Here, we review these proteomic methods and their application to studying viral and bacterial intracellular pathogens. We examine approaches for defining spatial and temporal host–pathogen protein interactions upon infection of a host cell. Further expanding the understanding of proteome organization during an infection, we discuss methods that characterize the regulation of host and pathogen proteomes through alterations in protein abundance, localization, and post‐translational modifications. Finally, we highlight bioinformatic tools available for analyzing such proteomic datasets, as well as novel strategies for integrating proteomics with other omic tools, such as genomics, transcriptomics, and metabolomics, to obtain a systems‐level understanding of infectious diseases.
Collapse
Affiliation(s)
- Pierre M Jean Beltran
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Joel D Federspiel
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Xinlei Sheng
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| |
Collapse
|
26
|
Current Perspectives on Cytomegalovirus in Heart Transplantation. CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Wang L, Li F, Dang L, Liang C, Wang C, He B, Liu J, Li D, Wu X, Xu X, Lu A, Zhang G. In Vivo Delivery Systems for Therapeutic Genome Editing. Int J Mol Sci 2016; 17:E626. [PMID: 27128905 PMCID: PMC4881452 DOI: 10.3390/ijms17050626] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/11/2016] [Accepted: 04/14/2016] [Indexed: 12/28/2022] Open
Abstract
Therapeutic genome editing technology has been widely used as a powerful tool for directly correcting genetic mutations in target pathological tissues and cells to cure of diseases. The modification of specific genomic sequences can be achieved by utilizing programmable nucleases, such as Meganucleases, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the clustered regularly-interspaced short palindromic repeat-associated nuclease Cas9 (CRISPR/Cas9). However, given the properties, such as large size, negative charge, low membrane penetrating ability, as well as weak tolerance for serum, and low endosomal escape, of these nucleases genome editing cannot be successfully applied unless in vivo delivery of related programmable nucleases into target organisms or cells is achieved. Here, we look back at delivery strategies having been used in the in vivo delivery of three main genome editing nucleases, followed by methodologies currently undergoing testing in clinical trials, and potential delivery strategies provided by analyzing characteristics of nucleases and commonly used vectors.
Collapse
Affiliation(s)
- Luyao Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Fangfei Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Lei Dang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Chao Liang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Chao Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Bing He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Defang Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Xiaohao Wu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Xuegong Xu
- Central Laboratory, Zheng Zhou Hospital of Traditional Chinese Medicine, Zhengzhou 450000, China.
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 00852, China.
| |
Collapse
|
28
|
Li L, He ZY, Wei XW, Gao GP, Wei YQ. Challenges in CRISPR/CAS9 Delivery: Potential Roles of Nonviral Vectors. Hum Gene Ther 2016; 26:452-62. [PMID: 26176432 DOI: 10.1089/hum.2015.069] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
CRISPR/Cas9 genome editing platforms are widely applied as powerful tools in basic research and potential therapeutics for genome regulation. The appropriate alternative of delivery system is critical if genome editing systems are to be effectively performed in the targeted cells or organisms. To date, the in vivo delivery of the Cas9 system remains challenging. Both physical methods and viral vectors are adopted in the delivery of the Cas9-based gene editing platform. However, physical methods are more applicable for in vitro delivery, while viral vectors are generally concerned with safety issues, limited packing capacities, and so on. With the robust development of nonviral drug delivery systems, lipid- or polymer-based nanocarriers might be potent vectors for the delivery of CRISPR/Cas9 systems. In this review, we look back at the delivery approaches that have been used for the delivery of the Cas9 system and outline the recent development of nonviral vectors that might be potential carriers for the genome editing platform in the future. The efforts in optimizing cationic nanocarriers with structural modification are described and promising nonviral vectors under clinical investigations are highlighted.
Collapse
Affiliation(s)
- Ling Li
- 1 Lab for Aging Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan, PR China
| | - Zhi-Yao He
- 1 Lab for Aging Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan, PR China
| | - Xia-Wei Wei
- 1 Lab for Aging Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan, PR China
| | - Guang-Ping Gao
- 2 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,3 Department of Microbiology and Physiology Systems, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Yu-Quan Wei
- 1 Lab for Aging Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan, PR China
| |
Collapse
|
29
|
Tabaei S, Mashkani B, Esmaili A, Karimi R, Jamehdar SA. Design of cocktail peptide vaccine against Cytomegalovirus infection. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:449-54. [PMID: 27279990 PMCID: PMC4887719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVES Human Cytomegalovirus (HCMV) remains a major morbidity and mortality cause in immuno suppressed patients. Therefore, significant effort has been made towards the development of a vaccine. In this study, the expression of the pp65 and gB fusion peptides and Fc domain of mouse IgG2a as a novel delivery system for selective uptake of antigens by antigen-presenting cells (APCs) in Pichia pastoris yeast system were studied. MATERIALS AND METHOD In this study, four immune dominant sequences in pp65 protein and 3 immuno dominant sequences in gB protein were selected according to literature review. Peptide linker -GGGGS- was used for construction of fusion peptide. This fusion peptide was cloned in the pPICZαA expression vector and transfected into P. pastoris host cells. RESULTS Dot blot and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) techniques showed that a high level of pp65-gB-Fc fusion peptide was expressed. CONCLUSION This CMV pp65-gB-Fc fusion peptide could be a promising candidate for the development of a novel peptide vaccine.
Collapse
Affiliation(s)
- Samira Tabaei
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Baratali Mashkani
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Esmaili
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Reza Karimi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Saeid Amel Jamehdar
- Antimicrobial Resistance Research Center, Avicenna Research Institute, Mashhad University of Medical Science, Mashhad, Iran,Corresponding author: Saeid Amel Jamehdar. Antimicrobial Resistance Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran. Tel: +98-51-3802 2205; Fax: +98-51-3711 2596;
| |
Collapse
|
30
|
Affiliation(s)
- Kristy M. Bialas
- Human Vaccine Institute, Duke University Medical School, Durham, North Carolina, United States of America
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University Medical School, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical School, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
31
|
Bernstein DI, Munoz FM, Callahan ST, Rupp R, Wootton SH, Edwards KM, Turley CB, Stanberry LR, Patel SM, Mcneal MM, Pichon S, Amegashie C, Bellamy AR. Safety and efficacy of a cytomegalovirus glycoprotein B (gB) vaccine in adolescent girls: A randomized clinical trial. Vaccine 2015; 34:313-9. [PMID: 26657184 DOI: 10.1016/j.vaccine.2015.11.056] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/20/2015] [Accepted: 11/21/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) is a leading cause of congenital infection and an important target for vaccine development. METHODS CMV seronegative girls between 12 and 17 years of age received CMV glycoprotein B (gB) vaccine with MF59 or saline placebo at 0, 1 and 6 months. Blood and urine were collected throughout the study for evidence of CMV infection based on PCR and/or seroconversion to non-vaccine CMV antigens. RESULTS 402 CMV seronegative subjects were vaccinated (195 vaccine, 207 placebo). The vaccine was generally well tolerated, although local and systemic adverse events were significantly more common in the vaccine group. The vaccine induced gB antibody in all vaccine recipients with a gB geometric mean titer of 13,400 EU; 95%CI 11,436, 15,700, after 3 doses. Overall, 48 CMV infections were detected (21 vaccine, 27 placebo). In the per protocol population (124 vaccine, 125 placebo) vaccine efficacy was 43%; 95%CI: -36; 76, p=0.20. The most significant difference was after 2 doses, administered as per protocol; vaccine efficacy 45%, 95%CI: -9; 72, p=0.08. CONCLUSION The vaccine was safe and immunogenic. Although the efficacy did not reach conventional levels of significance, the results are consistent with a previous study in adult women (Pass et al. N Engl J Med 2009;360:1191) using the same formulation.
Collapse
Affiliation(s)
- David I Bernstein
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States.
| | - Flor M Munoz
- Baylor College of Medicine, Houston, TX, United States
| | - S Todd Callahan
- Vanderbilt University Medical Center, Nashville, TN, United States
| | - Richard Rupp
- University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Susan H Wootton
- University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Christine B Turley
- University of Texas Medical Branch at Galveston, Galveston, TX, United States; University of South Carolina School of Medicine, Columbia, SC, United States
| | - Lawrence R Stanberry
- University of Texas Medical Branch at Galveston, Galveston, TX, United States; Columbia University, New York Presbyterian Hospital, New York City, NY, United States
| | | | - Monica M Mcneal
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | | | | | | |
Collapse
|
32
|
Awareness of Cytomegalovirus Infection among Pregnant Women in Geneva, Switzerland: A Cross-sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:15285-97. [PMID: 26633451 PMCID: PMC4690918 DOI: 10.3390/ijerph121214982] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cytomegalovirus (CMV) is the most frequent cause of congenital infection and commonly associated with sensorineural deficit. At present, there is neither prophylaxis nor treatment during pregnancy. The objective of this study was to evaluate the level of awareness regarding CMV infection and its consequences in women delivering at the University of Geneva Hospitals (Geneva, Switzerland). METHODS The study consisted of a validated questionnaire completed by women in the immediate postpartum period. RESULTS The questionnaire was completed by 59% (314/528) of delivering women. Only 39% (123/314) knew about CMV and 19.7% (62/314) had received information about preventive measures. Women were more aware about other congenital diseases, such as toxoplasmosis (87%); human immunodeficiency virus (99%); syphilis (85.5%); rubella (92.3%); and group B Streptococcus (63%). Factors associated with CMV awareness were Swiss nationality, high education level, employment in health care or with children, and being followed by an obstetrician. Regarding quality of information, few were aware of the main CMV complications (deafness, 25.2%; mental retardation, 34.5%). Among those informed about CMV, most (74.6%) knew about preventive measures. Among these, 82.5% thought that these were easily applicable. CONCLUSIONS Most women were unaware of CMV infection and its potential risks during pregnancy. It is crucial to improve CMV information given to pregnant women to prevent the risks for the fetus/newborn.
Collapse
|
33
|
Understanding factors influencing vaccination acceptance during pregnancy globally: A literature review. Vaccine 2015; 33:6420-9. [DOI: 10.1016/j.vaccine.2015.08.046] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/06/2015] [Accepted: 08/11/2015] [Indexed: 01/22/2023]
|
34
|
Rogers N. A dormant danger: New therapies target a ubiquitous pathogen known as cytomegalovirus. Nat Med 2015; 21:1104-5. [PMID: 26444624 DOI: 10.1038/nm1015-1104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Chandramouli S, Ciferri C, Nikitin PA, Caló S, Gerrein R, Balabanis K, Monroe J, Hebner C, Lilja AE, Settembre EC, Carfi A. Structure of HCMV glycoprotein B in the postfusion conformation bound to a neutralizing human antibody. Nat Commun 2015; 6:8176. [PMID: 26365435 PMCID: PMC4579600 DOI: 10.1038/ncomms9176] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/27/2015] [Indexed: 12/23/2022] Open
Abstract
Human cytomegalovirus (HCMV) poses a significant threat to immunocompromised individuals and neonates infected in utero. Glycoprotein B (gB), the herpesvirus fusion protein, is a target for neutralizing antibodies and a vaccine candidate due to its indispensable role in infection. Here we show the crystal structure of the HCMV gB ectodomain bound to the Fab fragment of 1G2, a neutralizing human monoclonal antibody isolated from a seropositive subject. The gB/1G2 interaction is dominated by aromatic residues in the 1G2 heavy chain CDR3 protruding into a hydrophobic cleft in the gB antigenic domain 5 (AD-5). Structural analysis and comparison with HSV gB suggest the location of additional neutralizing antibody binding sites on HCMV gB. Finally, immunoprecipitation experiments reveal that 1G2 can bind to HCMV virion gB suggesting that its epitope is exposed and accessible on the virus surface. Our data will support the development of vaccines and therapeutic antibodies against HCMV infection. Cytomegalovirus is a danger to individuals with compromised immune systems and neonates infected in utero. Here the authors show the structure of a neutralizing antibody-bound viral fusion protein glycoprotein B, supporting the development of therapeutic antibodies and vaccines.
Collapse
Affiliation(s)
| | - Claudio Ciferri
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - Pavel A Nikitin
- Novartis Institutes for Biomedical Research, Emeryville, California 94608, USA
| | - Stefano Caló
- GSK Vaccines, Via Fiorentina 1, Siena 53100, Italy
| | - Rachel Gerrein
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - Kara Balabanis
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - James Monroe
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - Christy Hebner
- Novartis Institutes for Biomedical Research, Emeryville, California 94608, USA
| | - Anders E Lilja
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - Ethan C Settembre
- Novartis Influenza Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| | - Andrea Carfi
- GSK Vaccines, 45 Sidney Street, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
36
|
A site of varicella-zoster virus vulnerability identified by structural studies of neutralizing antibodies bound to the glycoprotein complex gHgL. Proc Natl Acad Sci U S A 2015; 112:6056-61. [PMID: 25918416 PMCID: PMC4434712 DOI: 10.1073/pnas.1501176112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Mapping neutralizing epitopes on viral entry glycoproteins allows the identification of potentially important functional regions. The structure of varicella-zoster virus (VZV) gHgL bound to two antibodies isolated from immune donors reveals a common binding site. Functional experiments demonstrate that the two antibodies neutralize VZV infection and inhibit glycoprotein gB/glycoprotein complex gHgL-mediated membrane fusion. Immunization experiments in mice demonstrate that VZV gHgL elicits potently neutralizing antibodies and confirm the key role of this antigenic site in antibody-mediated virus neutralization. This manuscript sheds light on the molecular mechanism of herpesvirus cell entry and will guide the design of subunit-based vaccines against VZV. Varicella-zoster virus (VZV), of the family Alphaherpesvirinae, causes varicella in children and young adults, potentially leading to herpes zoster later in life on reactivation from latency. The conserved herpesvirus glycoprotein gB and the heterodimer gHgL mediate virion envelope fusion with cell membranes during virus entry. Naturally occurring neutralizing antibodies against herpesviruses target these entry proteins. To determine the molecular basis for VZV neutralization, crystal structures of gHgL were determined in complex with fragments of antigen binding (Fabs) from two human monoclonal antibodies, IgG-94 and IgG-RC, isolated from seropositive subjects. These structures reveal that the antibodies target the same site, composed of residues from both gH and gL, distinct from two other neutralizing epitopes identified by negative-stain electron microscopy and mutational analysis. Inhibition of gB/gHgL-mediated membrane fusion and structural comparisons with herpesvirus homologs suggest that the IgG-RC/94 epitope is in proximity to the site on VZV gHgL that activates gB. Immunization studies proved that the anti-gHgL IgG-RC/94 epitope is a critical target for antibodies that neutralize VZV. Thus, the gHgL/Fab structures delineate a site of herpesvirus vulnerability targeted by natural immunity.
Collapse
|
37
|
Wiegers AK, Sticht H, Winkler TH, Britt WJ, Mach M. Identification of a neutralizing epitope within antigenic domain 5 of glycoprotein B of human cytomegalovirus. J Virol 2015; 89:361-72. [PMID: 25320309 PMCID: PMC4301166 DOI: 10.1128/jvi.02393-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/07/2014] [Indexed: 01/19/2023] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is an important, ubiquitous pathogen that causes severe clinical disease in immunocompromised individuals, such as organ transplant recipients and infants infected in utero. The envelope glycoprotein B (gB) of HCMV is a major antigen for the induction of virus-neutralizing antibodies. We have begun to define target structures within gB that are recognized by virus-neutralizing antibodies. Antigenic domain 5 (AD-5) of gB has been identified as an important target for neutralizing antibodies in studies using human monoclonal antibodies (MAbs). Anti-AD-5 MAbs share a target site on gB, despite originating from different, healthy, HCMV-infected donors. Mutational analysis of AD-5 identified tyrosine 280 in combination with other surface-exposed residues (the YNND epitope) as critical for antibody binding. The YNND epitope is strictly conserved among different HCMV strains. Recombinant viruses carrying YNND mutations in AD-5 were resistant to virus-neutralizing MAbs. Competition enzyme-linked immunosorbent assays (ELISAs) with human HCMV-convalescent-phase sera from unselected donors confirmed the conserved antibody response for the YNND epitope in HCMV-infected individuals and, because a significant fraction of the gB AD-5 response was directed against the YNND epitope, further argued that this epitope is a major target of anti-AD-5 antibody responses. In addition, affinity-purified polyclonal anti-AD-5 antibodies prepared from individual sera showed reactivity to AD-5 and neutralization activity toward gB mutant viruses that were similar to those of AD-5-specific MAbs. Taken together, our data indicate that the YNND epitope represents an important target for anti-gB antibody responses as well as for anti-AD-5 virus-neutralizing antibodies. IMPORTANCE HCMV is a major global health concern, and a vaccine to prevent HCMV disease is a widely recognized medical need. Glycoprotein B of HCMV is an important target for neutralizing antibodies and hence an interesting molecule for intervention strategies, e.g., vaccination. Mapping the target structures of neutralizing antibodies induced by naturally occurring HCMV infection can aid in defining the properties required for a protective capacity of vaccine antigens. The data presented here extend our knowledge of neutralizing epitopes within gB to include AD-5. Collectively, our data will contribute to optimal vaccine design and development of antibody-based therapies.
Collapse
Affiliation(s)
- Anna-Katharina Wiegers
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Institut für Biochemie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H Winkler
- Nikolaus-Fiebiger-Zentrum für Molekulare Medizin, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - William J Britt
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael Mach
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
38
|
Huang C, Wang H, Wu S, Chang H, Liu L, Peng B, Fang F, Chen Z. Comparison of multiple DNA vaccines for protection against cytomegalovirus infection in BALB/c mice. Virol J 2014; 11:104. [PMID: 24898886 PMCID: PMC4073173 DOI: 10.1186/1743-422x-11-104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/23/2014] [Indexed: 11/23/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) causes serious HCMV-related diseases in immunocompromised people. Vaccination is the most effective measure to control infection with the pathogen, yet no vaccine has been licensed till now. We performed a head-to-head comparison of the protective abilities of multiple DNA vaccines in murine model of murine cytomegalovirus (MCMV) infection. Methods Five DNA vaccines were constructed. Four encoding MCMV proteins gp34 (m04), p65 (M84), DNA helicase (M105), and immediate-early 1 protein pp89 (IE-1) , respectively, which were reported to induce CD8+ T cell responses, were compared with the one expressing gB (M55), the neutralizing antibody target antigen, for immune protection in BALB/c mice. Mice were immunized with these DNA vaccines 1 to 4 times via intramuscular injection followed by electroporation, and were subsequently infected with a lethal dose (3 × LD50) of highly virulent SG-MCMV. Specific antibodies and IFN-γ secreting splenocytes were detected by immunoblotting and ELISPOT, respectively. Protective abilities in mice provided by the vaccines were evaluated by residual virus titers in organs, survival rate and weight loss. Results These DNA vaccines, especially m04, M84 and IE-1, could effectively reduce the virus loads in salivary glands and spleens of mice, but they couldn’t completely clear the residual virus. Survival rates of 100% in mice after a lethal dose of MCMV infection could be reached by more than one dose of M84 vaccine or two doses of m04 or IE-1 vaccine. Immunization with M55 or M105 DNA at four doses offered mice only 62.5% survival rate after the lethal challenge. Conclusions The study demonstrated that DNA vaccines could effectively afford mice protection against infection with a highly virulent MCMV and that the protection offered by induced CD8+ T cell immunity might be superior to that by gB-specific antibodies. These results are valuable references for development and application of HCMV vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fang Fang
- College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | | |
Collapse
|