1
|
Acharya A, Byrareddy SN. Immunological insights into the re-emergence of human metapneumovirus. Curr Opin Immunol 2025; 94:102562. [PMID: 40359650 PMCID: PMC12166946 DOI: 10.1016/j.coi.2025.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Human metapneumovirus (hMPV) is a seasonal respiratory virus that typically causes mild, flu-like symptoms. In some cases, it can lead to severe respiratory complications, such as pneumonia, bronchitis, and bronchiolitis, often requiring hospitalization. Recently, a surge in hMPV cases has been reported in China and other countries, raising concerns about a potential pandemic scenario reminiscent of COVID-19. This review explores the genomic structure, replication cycle, genetic diversity, and evolutionary trajectory of hMPV. It also discusses host immune responses and the available animal models to study pathogenesis and to screen for potential vaccines and antivirals. Additionally, we examine the shifting seasonal trends in hMPV circulation, evaluate the low pandemic risk posed by existing hMPV clades, and underscore the need for continued vaccine and antiviral development. Finally, we advocate for strengthened global surveillance, especially in low- and middle-income countries, as a critical strategy to mitigate the risks posed by emerging hMPV clades.
Collapse
Affiliation(s)
- Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
2
|
Wilkhoo HS, Islam AW, Wilkhoo HS, Hussain S, Singh B, Kadam SR. Clinical Insights and Advancements in Human Metapneumovirus Management and Prognosis. Discoveries (Craiova) 2025; 13:e204. [PMID: 40351504 PMCID: PMC12061465 DOI: 10.15190/d.2025.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Human metapneumovirus is a respiratory pathogen that infects children, the elderly, and immunocompromised individuals. Despite its global prevalence, underdiagnosis persists because of clinical overlap with other respiratory viruses. The current approach is mostly supportive, with oxygen therapy and hydration being crucial interventions. Ribavirin contains antiviral properties but has little clinical application. Vaccine development is moving forward, with prospects including live-attenuated, subunit-based, and virus-like particle vaccines. Molecular diagnostics, such as RT-PCR and metagenomic sequencing, have increased detection rates, which aids epidemiological monitoring. Monoclonal antibodies targeting the fusion (F) protein are being studied for passive immunity, while immunomodulatory treatments such as corticosteroids and intravenous immunoglobulins may help treat severe cases. Emerging treatments include fusion inhibitors and pan-pneumovirus vaccinations that protect against HMPV and RSV. Future research should concentrate on optimizing antiviral methods, increasing vaccination trials, and improving surveillance to detect outbreaks. A multidisciplinary approach that combines virology, immunology, and epidemiology is required to reduce HMPV's effect and improve patient outcomes. This review serves as a comprehensive literature about HMPV which provides all the crucial clinical perspectives and the latest advancements in management, antivirals, patient prognosis as well and diagnostic modalities.
Collapse
Affiliation(s)
- Harsahaj Singh Wilkhoo
- Faculty of Medicine, Tbilisi State Medical University, Tbilisi, Georgia
- ClinNova International, Tbilisi, Georgia
| | - Afra Wasama Islam
- Faculty of Medicine, Tbilisi State Medical University, Tbilisi, Georgia
- ClinNova International, Tbilisi, Georgia
| | | | - Suhaib Hussain
- Faculty of Medicine, Tbilisi State Medical University, Tbilisi, Georgia
- ClinNova International, Tbilisi, Georgia
| | - Bharat Singh
- Faculty of Medicine, Tbilisi State Medical University, Tbilisi, Georgia
| | | |
Collapse
|
3
|
Koutsoumpli G, Stasiukonyte N, Hoogeboom BN, Daemen T. An in vitro CD8 T-cell priming assay enables epitope selection for hepatitis C virus vaccines. Vaccine 2024; 42:126032. [PMID: 38964950 DOI: 10.1016/j.vaccine.2024.05.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024]
Abstract
For the rational design of epitope-specific vaccines, identifying epitopes that can be processed and presented is essential. As algorithm-based epitope prediction is frequently discordant with actually recognized CD8+ T-cell epitopes, we developed an in vitro CD8 T-cell priming protocol to enable the identification of truly and functionally expressed HLA class I epitopes. The assay was established and validated to identify epitopes presented by hepatitis C virus (HCV)-infected cells. In vitro priming of naïve CD8 T cells was achieved by culturing unfractionated PBMCs in the presence of a specific cocktail of growth factors and cytokines, and next exposing the cells to hepatic cells expressing the NS3 protein of HCV. After a 10-day co-culture, HCV-specific T-cell responses were identified based on IFN-γ ELISpot analysis. For this, the T cells were restimulated with long synthetic peptides (SLPs) spanning the whole NS3 protein sequence allowing the identification of HCV-specificity. We demonstrated that this protocol resulted in the in vitro priming of naïve precursors to antigen-experienced T-cells specific for 11 out of 98 SLPs tested. These 11 SLPs contain 12 different HLA-A*02:01-restricted epitopes, as predicted by a combination of three epitope prediction algorithms. Furthermore, we identified responses against 3 peptides that were not predicted to contain any immunogenic HLA class I epitopes, yet showed HCV-specific responses in vitro. Separation of CD8+ and CD8- T cells from PBMCs primed in vitro showed responses only upon restimulation with short peptides. We established an in vitro method that enables the identification of HLA class I epitopes resulting from cross-presented antigens and that can cross-prime T cells and allows the effective selection of functional immunogenic epitopes, but also less immunogenic ones, for the design of tailored therapeutic vaccines against persistent viral infections and tumor antigens.
Collapse
Affiliation(s)
- Georgia Koutsoumpli
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, PO Box 30 001, HPC EB88, 9700RB Groningen, the Netherlands
| | - Neringa Stasiukonyte
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, PO Box 30 001, HPC EB88, 9700RB Groningen, the Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, PO Box 30 001, HPC EB88, 9700RB Groningen, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, PO Box 30 001, HPC EB88, 9700RB Groningen, the Netherlands.
| |
Collapse
|
4
|
Sojati J, Zhang Y, Williams JV. Clinical human metapneumovirus isolates show distinct pathogenesis and inflammatory profiles but similar CD8 + T cell impairment. mSphere 2024; 9:e0057023. [PMID: 38197640 PMCID: PMC10826344 DOI: 10.1128/msphere.00570-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/01/2023] [Indexed: 01/11/2024] Open
Abstract
Human metapneumovirus (HMPV) is a negative-sense single-stranded RNA virus in the Pneumoviridae family and a leading cause of acute upper and lower respiratory infections, particularly in children, immunocompromised patients, and the elderly. Although nearly every person is infected with HMPV during early childhood, re-infections occur often, highlighting difficulty in building long-term immunity. Inflammatory responses, including PD-1-mediated impairment of virus-specific CD8+ T cells (TCD8), contribute to HMPV disease severity. HMPV strains are divided into four lineages: A1, A2, B1, and B2. However, little is known about immune responses to different viral subtypes. Here, we characterize responses to four HMPV clinical isolates-TN/94-344 (A1), TN/94-49 (A2), C2-202 (B1), and TN/96-35 (B2)-in vivo in C57BL/6 (B6) mice. TN/94-49 was avirulent, while TN/94-344, C2-202, and TN/96-35 showed varying degrees of weight loss and clinical disease. Differences in disease did not correlate to virus burden in upper or lower tracts. TN/94-49 HMPV exhibited highest nose titers and delayed lung clearance. Cytokine profiles differed between HMPV isolates, with TN/96-35 inducing the broadest lung inflammatory cytokines. TN/96-35 also showed lower HMPV burden and less weight loss than other virulent isolates, suggesting a more efficient antiviral response. Interestingly, disease correlated with higher expression of T-cell chemoattractant CXCL9. All isolates elicited PD-1 upregulation and decreased IFNγ and CD107a expression in virus-specific TCD8, with little difference between HMPV subtypes. This work uncovers previously uncharacterized variations in immune responses to clinical HMPV isolates of different lineages.IMPORTANCEThis study extensively explored differences in T-cell-mediated immunity between human metapneumovirus (HMPV) clinical isolates. Much existing HMPV research has been done with strains passaged extensively in cell lines, likely acquiring mutations advantageous to in vitro replication. Clinical isolates are collected directly from human patients and have undergone <10 passages, serving as more physiologically relevant models of HMPV infection. Additionally, existing animal studies of HMPV disease mainly focus on lung pathogenesis, while HMPV infects both upper and lower airways of humans. This work highlights distinct differences in HMPV burden in upper and lower tracts between clinical isolates. Lastly, this study uniquely explores differences in host immunity between all four HMPV genetic lineages. The predominant HMPV subtype in circulation varies seasonally; thus, understanding host responses to all subgroups is critical for developing effective HMPV vaccines.
Collapse
Affiliation(s)
- Jorna Sojati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Program in Microbiology & Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yu Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John V. Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute for Infection, Immunity, and Inflammation in Children, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Ma S, Zhu F, Xu Y, Wen H, Rao M, Zhang P, Peng W, Cui Y, Yang H, Tan C, Chen J, Pan P. Development of a novel multi-epitope mRNA vaccine candidate to combat HMPV virus. Hum Vaccin Immunother 2023; 19:2293300. [PMID: 38172569 PMCID: PMC10824151 DOI: 10.1080/21645515.2023.2293300] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Human metapneumovirus (HMPV) is one of the main pathogens causing severe respiratory infections in children, as a common cause of immunodeficiency-related deaths in children and elderly individuals, the prevalence of HMPV has been showing an increasing trend during the last years. However, no vaccines or effective treatment plans are available currently. In this present, based on candidate proteins highly associated with viral virulence and has promising protective potential, we screened for immunodominant cytotoxic T cells, helper T cells, and Linear B-cell epitopes from the most promising candidate Fusion protein, together with G, SH, M, and M2. All epitopes were predicted to have strong antigenicity by Vaxijen and pose no potential toxicity, allergenicity, or hormonology to human proteins by Toxinpred, Allerpred, and Blast analysis, meanwhile, high conservancy is demanded to cover different subtypes. adjuvants β-defensin II and Pam2Cys was attached with EAAAK linkers to improve vaccine's efficiency. Then, calculation of physicochemical properties proved the protein vaccine as a product can stably exist in the human body. Besides, we assessed the docking between the vaccine and immune receptors to evaluate its ability to stimulate immune responses, and the dynamic simulation further confirmed that the vaccine can tightly bind with immune receptors, which approved that the construction has the potential to induce strong humoral and cellular immune response. Finally, the vaccine was constructed into a multi-epitope mRNA vaccine, the immune simulations suggest that this is a vaccine candidate for controlling HMPV infection.
Collapse
Affiliation(s)
- Shiyang Ma
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Fei Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Yizhong Xu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Haicheng Wen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Mingjun Rao
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Peipei Zhang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Wenzhong Peng
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Yanhui Cui
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Hang Yang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Caixia Tan
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
6
|
Chongyu T, Guanglin L, Fang S, Zhuoya D, Hao Y, Cong L, Xinyu L, Wei H, Lingyun T, Yan N, Penghui Y. A chimeric influenza virus vaccine expressing fusion protein epitopes induces protection from human metapneumovirus challenge in mice. Front Microbiol 2023; 13:1012873. [PMID: 38155756 PMCID: PMC10753001 DOI: 10.3389/fmicb.2022.1012873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/19/2022] [Indexed: 12/30/2023] Open
Abstract
Human metapneumovirus (HMPV) is a common virus associated with acute respiratory distress syndrome in pediatric patients. There are no HMPV vaccines or therapeutics that have been approved for prevention or treatment. In this study, we constructed a novel recombinant influenza virus carrying partial HMPV fusion protein (HMPV-F), termed rFLU-HMPV/F-NS, utilizing reverse genetics, which contained (HMPV-F) in the background of NS segments of influenza virus A/PuertoRico/8/34(PR8). The morphological characteristics of rFLU-HMPV/F-NS were consistent with the wild-type flu virus. Additionally, immunofluorescence results showed that fusion proteins in the chimeric rFLU-HMPV/F-NS could work well, and the virus could be stably passaged in SPF chicken embryos. Furthermore, intranasal immunization with rFLU-HMPV/F-NS in BALB/c mice induced robust humoral, mucosal and Th1-type dominant cellular immune responses in vivo. More importantly, we discovered that rFLU-HMPV/F-NS afforded significant protective efficacy against the wild-type HMPV and influenza virus challenge, with significantly attenuated pathological changes and reduced viral titers in the lung tissues of immunized mice. Collectively, these findings demonstrated that chimeric recombinant rFLU-HMPV/F-NS as a promising HMPV candidate vaccine has potentials for the development of HMPV vaccine.
Collapse
Affiliation(s)
- Tian Chongyu
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Lei Guanglin
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Sun Fang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Deng Zhuoya
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Hao
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li Cong
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li Xinyu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - He Wei
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Tan Lingyun
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Niu Yan
- Inner Mongolia Medical University, Hohhot, China
| | - Yang Penghui
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Inner Mongolia Medical University, Hohhot, China
- First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Miranda-Katz M, Erickson JJ, Lan J, Ecker A, Zhang Y, Joyce S, Williams JV. Novel HLA-B7-restricted human metapneumovirus epitopes enhance viral clearance in mice and are recognized by human CD8 + T cells. Sci Rep 2021; 11:20769. [PMID: 34675220 PMCID: PMC8531189 DOI: 10.1038/s41598-021-00023-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/05/2021] [Indexed: 11/09/2022] Open
Abstract
Human metapneumovirus (HMPV) is a leading cause of acute lower respiratory tract illness in children and adults. Repeated infections are common and can be severe in young, elderly, and immunocompromised persons due to short-lived protective humoral immunity. In turn, few protective T cell epitopes have been identified in humans. Thus, we infected transgenic mice expressing the common human HLA MHC-I allele B*07:02 (HLA-B7) with HMPV and screened a robust library of overlapping and computationally predicted HLA-B7 binding peptides. Six HLA-B7-restricted CD8+ T cell epitopes were identified using ELISPOT screening in the F, M, and N proteins, with M195-203 (M195) eliciting the strongest responses. MHC-tetramer flow cytometric staining confirmed HLA-B7 epitope-specific CD8+ T cells migrated to lungs and spleen of HMPV-immune mice. Immunization with pooled HLA-B7-restricted peptides reduced viral titer and protected mice from virulent infection. Finally, we confirmed that CD8+ T cells from HLA-B7 positive humans also recognize the identified epitopes. These results enable identification of HMPV-specific CD8+ T cells in humans and help to inform future HMPV vaccine design.
Collapse
Affiliation(s)
- Margot Miranda-Katz
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave, Rangos 9122, Pittsburgh, PA, 15224, USA
| | - John J Erickson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, USA
| | - Jie Lan
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave, Rangos 9122, Pittsburgh, PA, 15224, USA
| | - Alwyn Ecker
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave, Rangos 9122, Pittsburgh, PA, 15224, USA
| | - Yu Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave, Rangos 9122, Pittsburgh, PA, 15224, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, USA
- Vanderbilt Institute for Infection, Immunity, and Inflammation (VI4), Nashville, TN, 37232, USA
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave, Rangos 9122, Pittsburgh, PA, 15224, USA.
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, PA, 15224, USA.
| |
Collapse
|
8
|
Choi EJ, Wu W, Chen Y, Yan W, Li L, Choudhury A, Bao X. The role of M2-2 PDZ-binding motifs in pulmonary innate immune responses to human metapneumovirus. J Med Virol 2020; 92:2946-2954. [PMID: 32073159 PMCID: PMC8357536 DOI: 10.1002/jmv.25713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
Human metapneumovirus (HMPV) is a leading cause of lower respiratory tract infection (LRTI) in pediatric and geriatric populations. We recently found that two PDZ-binding motifs of the M2-2 protein, 29-DEMI-32 and 39-KEALSDGI-46, play a significant role in mediating HMPV immune evasion in airway epithelial cells (AECs). However, their role in the overall pulmonary responses to HMPV infection has not been investigated. In this study, we found that two recombinant HMPVs (rHMPV) lacking the individual M2-2 PDZ-binding motif are attenuated in mouse lungs. Mice infected with mutants produce more cytokines/chemokines in bronchoalveolar lavage (BAL) fluid compared to mice infected with wild-type rHMPV. In addition, both mutants are able to enhance the pulmonary recruitment of dendritic cells (DCs) and T cells and induce effective protections against the HMPV challenge. The DC maturation is also significantly improved by the motif mutation. Taken together, our data provide proof-of-principle for two live-attenuated M2-2 mutants to be promising HMPV vaccine candidates that are effective in inducing higher pulmonary innate immunity and generating protection against HMPV infection.
Collapse
Affiliation(s)
- Eun-Jin Choi
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Yu Chen
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Weiyu Yan
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Honeybee Research Institute, Jiangxi Agriculture University, Nanchang, Jiangxi, China
| | - Liqing Li
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Department of Microbiology, The University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Atanu Choudhury
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- The University of Texas at Austin, Austin, TX78712, USA
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX 77555, USA
- The Institute of Translational Sciences, UTMB, Galveston, TX 77555, USA
- The Institute for Human Infections and Immunity, UTMB, Galveston, TX 77555, USA
| |
Collapse
|
9
|
Diab M, Schmiedel D, Seidel E, Bacharach E, Mandelboim O. Human Metapneumovirus Escapes NK Cell Recognition through the Downregulation of Stress-Induced Ligands for NKG2D. Viruses 2020; 12:v12070781. [PMID: 32698530 PMCID: PMC7412239 DOI: 10.3390/v12070781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 11/28/2022] Open
Abstract
The Pneumoviridae family includes human metapneumovirus (HMPV) and human orthopneumovirus, which is also known as a respiratory syncytial virus (HRSV). These are large enveloped, negative single-strand RNA viruses. HMPV and HRSV are the human members, which commonly infect children. HMPV, which was discovered in 2001, infects most children until the age of five, which causes an influenza-like illness. The interaction of this virus with immune cells is poorly understood. In this study, we show that HMPV evades natural killer (NK) cell attack by downregulating stress-induced ligands for the activating receptor NKG2D including: Major histocompatibility complex (MHC) class I polypeptide-related sequences A and B (MICA, MICB), UL16 binding proteins ULBP2, and ULBP3, but not ULBP1. Mechanistically, we show that the viral protein G is involved in the downregulation of ULBP2 and that the viral protein M2.2 is required for MICA and MICB downregulation. These findings emphasize the importance of NK cells, in general, and NKG2D, in particular, in controlling HMPV infection, which opens new avenues for treating HMPV.
Collapse
Affiliation(s)
- Mohammad Diab
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Dominik Schmiedel
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Einat Seidel
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Eran Bacharach
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
- Correspondence: ; Tel.: +972-2675-7515
| |
Collapse
|
10
|
Prospects of and Barriers to the Development of Epitope-Based Vaccines against Human Metapneumovirus. Pathogens 2020; 9:pathogens9060481. [PMID: 32570728 PMCID: PMC7350342 DOI: 10.3390/pathogens9060481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory illnesses in children, the elderly and immunocompromised patients. Although this pathogen was only discovered in 2001, an enormous amount of research has been conducted in order to develop safe and effective vaccines to prevent people from contracting the disease. In this review, we summarize current knowledge about the most promising experimental B- and T-cell epitopes of human metapneumovirus for the rational design of HMPV vaccines using vector delivery systems, paying special attention to the conservation of these epitopes among different lineages/genotypes of HMPV. The prospects of the successful development of an epitope-based HMPV vaccine are discussed in the context of recent findings regarding HMPV’s ability to modulate host immunity. In particular, we discuss the lack of data on experimental human CD4 T-cell epitopes for HMPV despite the role of CD4 lymphocytes in both the induction of higher neutralizing antibody titers and the establishment of CD8 memory T-cell responses. We conclude that current research should be focused on searching for human CD4 T-cell epitopes of HMPV that can help us to design a safe and cross-protective epitope-based HMPV vaccine.
Collapse
|
11
|
Kumar P, Srivastava M. Prophylactic and therapeutic approaches for human metapneumovirus. Virusdisease 2018; 29:434-444. [PMID: 30539045 PMCID: PMC6261883 DOI: 10.1007/s13337-018-0498-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Human metapneumovirus (HMPV) is an important pneumovirus which causes acute respiratory disease in human beings. The viral infection leads to mild to severe respiratory symptoms depending on the age and immune status of the infected individual. Several groups across the world are working on the development of immunogens and therapy to manage HMPV infection with promising results under laboratory conditions but till date any virus specific vaccine or therapy has not been approved for clinical use. This minireview gives an overview of the prophylactic and therapeutic approaches to manage HMPV infections.
Collapse
Affiliation(s)
- Prashant Kumar
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, U.P. 201301 India
| | - Mansi Srivastava
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, U.P. 201301 India
| |
Collapse
|
12
|
Marsico S, Caccuri F, Mazzuca P, Apostoli P, Roversi S, Lorenzin G, Zani A, Fiorentini S, Giagulli C, Caruso A. Human lung epithelial cells support human metapneumovirus persistence by overcoming apoptosis. Pathog Dis 2018; 76:4923026. [PMID: 29617859 DOI: 10.1093/femspd/fty013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/05/2018] [Indexed: 11/12/2022] Open
Abstract
Human metapneumovirus (hMPV) has been identified as a major cause of lower respiratory tract infection in children. Epidemiological and molecular evidence has highlighted an association between severe childhood respiratory viral infection and chronic lung diseases, such as asthma and chronic obstructive pulmonary disease. Currently, animal models have demonstrated the ability of hMPV to persist in vivo suggesting a role of the virus in asthma development in children. However, mechanisms involved in hMPV persistence in the respiratory tract are not yet understood. In the present study we monitored hMPV infection in human alveolar epithelial A549 cells in order to understand if the virus is able to persist in these cells upon acute infection. Our data show that hMPV initially induces an apoptotic process in A549 cells through poly (ADP-ribose) polymerase 1 cleavage, caspase-3/7 activation and Wee1 activity. The hMPV-infected cells were then able to overcome the apoptotic pathway and cell cycle arrest in G2/M by expressing B-cell lymphoma 2 and to acquire a reservoir cell phenotype with constant production of infectious virus. These findings provide evidence of the ability of hMPV to persist in alveolar epithelial cells and help in understanding the mechanisms responsible for hMPV persistence in the human respiratory tract.
Collapse
Affiliation(s)
- Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Pietro Mazzuca
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Paola Apostoli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sara Roversi
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Giovanni Lorenzin
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Simona Fiorentini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Cinzia Giagulli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
13
|
Schmidt ME, Varga SM. The CD8 T Cell Response to Respiratory Virus Infections. Front Immunol 2018; 9:678. [PMID: 29686673 PMCID: PMC5900024 DOI: 10.3389/fimmu.2018.00678] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Humans are highly susceptible to infection with respiratory viruses including respiratory syncytial virus (RSV), influenza virus, human metapneumovirus, rhinovirus, coronavirus, and parainfluenza virus. While some viruses simply cause symptoms of the common cold, many respiratory viruses induce severe bronchiolitis, pneumonia, and even death following infection. Despite the immense clinical burden, the majority of the most common pulmonary viruses lack long-lasting efficacious vaccines. Nearly all current vaccination strategies are designed to elicit broadly neutralizing antibodies, which prevent severe disease following a subsequent infection. However, the mucosal antibody response to many respiratory viruses is not long-lasting and declines with age. CD8 T cells are critical for mediating clearance following many acute viral infections in the lung. In addition, memory CD8 T cells are capable of providing protection against secondary infections. Therefore, the combined induction of virus-specific CD8 T cells and antibodies may provide optimal protective immunity. Herein, we review the current literature on CD8 T cell responses induced by respiratory virus infections. Additionally, we explore how this knowledge could be utilized in the development of future vaccines against respiratory viruses, with a special emphasis on RSV vaccination.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
14
|
Abstract
Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection, particularly in children, immunocompromised patients, and the elderly. HMPV, which is closely related to avian metapneumovirus subtype C, has circulated for at least 65 years, and nearly every child will be infected with HMPV by the age of 5. However, immunity is incomplete, and re-infections occur throughout adult life. Symptoms are similar to those of other respiratory viral infections, ranging from mild (cough, rhinorrhea, and fever) to more severe (bronchiolitis and pneumonia). The preferred method for diagnosis is reverse transcription-polymerase chain reaction as HMPV is difficult to culture. Although there have been many advances made in the past 16 years since its discovery, there are still no US Food and Drug Administration-approved antivirals or vaccines available to treat HMPV. Both small animal and non-human primate models have been established for the study of HMPV. This review will focus on the epidemiology, transmission, and clinical manifestations in humans as well as the animal models of HMPV pathogenesis and host immune response.
Collapse
Affiliation(s)
- Nazly Shafagati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Diaz-Dinamarca DA, Ibañez FJ, Soto DA, Soto JA, Cespedes PF, Muena NA, Garate DS, Kalergis AM, Vasquez AE. Immunization with a Mixture of Nucleoprotein from Human Metapneumovirus and AbISCO-100 Adjuvant Reduces Viral Infection in Mice Model. Viral Immunol 2018; 31:306-314. [PMID: 29373084 DOI: 10.1089/vim.2017.0159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The human metapneumovirus (hMPV) is the second leading cause globally of acute infection of the respiratory tract in children, infecting the upper and lower airways. The hMPV may induce an inappropriate Th2-type immune response, which causes severe pulmonary inflammation, leading to the obstruction of airways. Despite its severe epidemiological relevance, no vaccines are currently available for the prevention of hMPV-induced illness. In this investigation, we demonstrated that immunization of mice with the recombinant hMPV nucleoprotein (hMPV-N) mixed with the AbISCO-100 adjuvant reduced viral replication in lungs following challenge with the virus. We found that immunized mice had reduced weight loss, decreased granulocytes in the lung, an increased level of specific nucleoprotein antibodies of IgG1 and IgG2a-isotypes, and a local profile of Th1/Th17-type cytokines. Our results suggest that immunization with the hMPV-N and the AbISCO-100 adjuvant induces a reduction of viral infection and could be considered for the development of an hMPV vaccine.
Collapse
Affiliation(s)
- Diego A Diaz-Dinamarca
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco J Ibañez
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel A Soto
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Jorge A Soto
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Cespedes
- 2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás A Muena
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Diego S Garate
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Alexis M Kalergis
- 2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile .,3 Facultad de Medicina, Departamento de Endocrinología, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Abel E Vasquez
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,4 Universidad San Sebastián , Facultad de Ciencia, Escuela de Bioquímica, Providencia, Santiago, Chile
| |
Collapse
|
16
|
Ren Y, Choi E, Zhang K, Chen Y, Ye S, Deng X, Zhang K, Bao X. Detection of Nuclear Protein Profile Changes by Human Metapneumovirus M2-2 Protein Using Quantitative Differential Proteomics. Vaccines (Basel) 2017; 5:vaccines5040045. [PMID: 29207503 PMCID: PMC5748611 DOI: 10.3390/vaccines5040045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 01/28/2023] Open
Abstract
Human metapneumovirus (hMPV) is a leading cause of lower respiratory infection in pediatric populations globally. This study examined proteomic profile changes in A549 cells infected with hMPV and two attenuated mutants with deleted PDZ domain-binding motif(s) in the M2-2 protein. These motifs are involved in the interruption of antiviral signaling, namely the interaction between the TNF receptor associated factor (TRAF) and mitochondrial antiviral-signaling (MAVS) proteins. The aim of this study was to provide insight into the overall and novel impact of M2-2 motifs on cellular responses via an unbiased comparison. Tandem mass tagging, stable isotope labeling, and high-resolution mass spectrometry were used for quantitative proteomic analysis. Using quantitative proteomics and Venn analysis, 1248 common proteins were detected in all infected samples of both technical sets. Hierarchical clustering of the differentiated proteome displayed distinct proteomic signatures that were controlled by the motif(s). Bioinformatics and experimental analysis confirmed the differentiated proteomes, revealed novel cellular biological events, and implicated key pathways controlled by hMPV M2-2 PDZ domain-binding motif(s). This provides further insight for evaluating M2-2 mutants as potent vaccine candidates.
Collapse
Affiliation(s)
- Yuping Ren
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Plastic Surgery, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan 430073, China.
| | - Eunjin Choi
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Ke Zhang
- Department of Biochemistry, Baylor University, Waco, TX 76706, USA.
| | - Yu Chen
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Pediatrics, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan 430073, China.
| | - Sha Ye
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Gynecologic Oncology Ward V, Hunan Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha 410008, China.
| | - Xiaoling Deng
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Xiaoyong Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.
- The Institute of Translational Science, University of Texas Medical Branch, Galveston, TX 77555, USA.
- The Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
17
|
Kan-O K, Ramirez R, MacDonald MI, Rolph M, Rudd PA, Spann KM, Mahalingam S, Bardin PG, Thomas BJ. Human Metapneumovirus Infection in Chronic Obstructive Pulmonary Disease: Impact of Glucocorticosteroids and Interferon. J Infect Dis 2017; 215:1536-1545. [PMID: 28379462 DOI: 10.1093/infdis/jix167] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
Background Human metapneumovirus (hMPV) infection is implicated in exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Research into the pathogenesis of infection is restricted to animal models, and information about hMPV replication and inflammatory and immune responses in human disease is limited. Methods Human primary bronchial epithelial cells (PBECs) from healthy and asthmatic subjects and those with COPD were infected with hMPV, with or without glucocorticosteroid (GCS) exposure. Viral replication, inflammatory and immune responses, and apoptosis were analyzed. We also determined whether adjuvant interferon (IFN) can blunt hMPV infection in vitro and in a murine model. Results hMPV infected human PBECs and viral replication was enhanced in cells from patients with COPD. The virus induced gene expression of IFN-stimulated gene 56 (ISG56) and IFN-β, as well as IFN-γ-inducible protein 10 (IP-10) and regulated on activation, normal T cell expressed and secreted (RANTES), and more so in cells from patients with COPD. GCS exposure enhanced hMPV replication despite increased IFN expression. Augmented virus replication associated with GCS was mediated by reduced apoptosis via induction of antiapoptotic genes. Adjuvant IFN treatment suppressed hMPV replication in PBECs and reduced hMPV viral titers and inflammation in vivo. Conclusions hMPV infects human PBECs, eliciting innate and inflammatory responses. Replication is enhanced by GCS and adjuvant IFN is an effective treatment, restricting virus replication and proinflammatory consequences of hMPV infections.
Collapse
Affiliation(s)
- Keiko Kan-O
- Monash Lung & Sleep, Monash Medical Centre.,Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, and.,Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne.,Research Institute for Disease of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | - Michael Rolph
- Institute for Glycomics, Griffith University, Southport, and
| | - Penny A Rudd
- Institute for Glycomics, Griffith University, Southport, and
| | - Kirsten M Spann
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | | | - Philip G Bardin
- Monash Lung & Sleep, Monash Medical Centre.,Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, and.,Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne
| | - Belinda J Thomas
- Monash Lung & Sleep, Monash Medical Centre.,Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, and.,Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne
| |
Collapse
|
18
|
Role of human metapneumovirus and respiratory syncytial virus in asthma exacerbations: where are we now? Clin Sci (Lond) 2017; 131:1713-1721. [PMID: 28667069 DOI: 10.1042/cs20160011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 04/18/2017] [Accepted: 05/02/2017] [Indexed: 12/30/2022]
Abstract
Since its discovery in 2001, human metapneumovirus (hMPV) has been identified as an important cause of respiratory tract infection in young children, second only to the closely related respiratory syncytial virus (RSV). Clinical evidence suggests that hMPV is associated with acute exacerbations of asthma in both children and adults, and may play a role in initiating asthma development in children. Animal models have demonstrated that airway hyperresponsiveness (AHR) and inflammation are triggered following hMPV infection, and hMPV is able to persist in vivo by inhibiting innate immune responses and causing aberrant adaptive responses. In this review, we discuss the prevalence of hMPV infection in pediatric and adult populations and its potential role in asthma exacerbation. We also review recent advances made in animal models to determine immune responses following hMPV infection, and compare to what is known about RSV.
Collapse
|
19
|
Abstract
Globally, as a leading agent of acute respiratory tract infections in children <5 years of age and the elderly, the human metapneumovirus (HMPV) has gained considerable attention. As inferred from studies comparing vaccinated and experimentally infected mice, the acquired immune response elicited by this pathogen fails to efficiently clear the virus from the airways, which leads to an exaggerated inflammatory response and lung damage. Furthermore, after disease resolution, there is a poor development of T and B cell immunological memory, which is believed to promote reinfections and viral spread in the community. In this article, we discuss the molecular mechanisms that shape the interactions of HMPV with host tissues that lead to pulmonary pathology and to the development of adaptive immunity that fails to protect against natural infections by this virus.
Collapse
|
20
|
Diab M, Glasner A, Isaacson B, Bar-On Y, Drori Y, Yamin R, Duev-Cohen A, Danziger O, Zamostiano R, Mandelboim M, Jonjic S, Bacharach E, Mandelboim O. NK-cell receptors NKp46 and NCR1 control human metapneumovirus infection. Eur J Immunol 2017; 47:692-703. [DOI: 10.1002/eji.201646756] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/20/2016] [Accepted: 02/08/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Mohammad Diab
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Ariella Glasner
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Batya Isaacson
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yotam Bar-On
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yaron Drori
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Rachel Yamin
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Alexandra Duev-Cohen
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Oded Danziger
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Rachel Zamostiano
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine; University of Rijeka; Rijeka Croatia
- Center for Proteomics, Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Eran Bacharach
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| |
Collapse
|
21
|
Márquez-Escobar VA. Current developments and prospects on human metapneumovirus vaccines. Expert Rev Vaccines 2017; 16:419-431. [PMID: 28116910 DOI: 10.1080/14760584.2017.1283223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Human metapneumovirus (hMPV) has become one of the major pathogens causing acute respiratory infections (ARI) mainly affecting young children, immunocompromised patients, and the elderly. Currently there are no licensed vaccines against this virus. Areas covered: Since the discovery of hMPV in 2001, many groups have focused on developing vaccines against this pathogen. This review presents the outcomes and perspectives derived from preclinical studies performed in cell cultures and animals as well as the only candidate that has reached evaluation in a clinical trial. Limitations of the current vaccine candidates are discussed and perspectives for the development of plant-based vaccines are analyzed. Expert commentary: Several hMPV vaccine candidates are under development with the potential to progress into clinical trials. In parallel, the molecular farming field offers new opportunities to generate innovative vaccines that will offer several advantages in the fight against hMPV.
Collapse
Affiliation(s)
- Verónica Araceli Márquez-Escobar
- a Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , Av. Dr. Manuel Nava 6, San Luis Potosí 78210 , SLP , Mexico
| |
Collapse
|
22
|
Gasper DJ, Neldner B, Plisch EH, Rustom H, Carrow E, Imai H, Kawaoka Y, Suresh M. Effective Respiratory CD8 T-Cell Immunity to Influenza Virus Induced by Intranasal Carbomer-Lecithin-Adjuvanted Non-replicating Vaccines. PLoS Pathog 2016; 12:e1006064. [PMID: 27997610 PMCID: PMC5173246 DOI: 10.1371/journal.ppat.1006064] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/14/2016] [Indexed: 01/31/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are critical for clearing many viral infections, and protective CTL memory can be induced by vaccination with attenuated viruses and vectors. Non-replicating vaccines are typically potentiated by the addition of adjuvants that enhance humoral responses, however few are capable of generating CTL responses. Adjuplex is a carbomer-lecithin-based adjuvant demonstrated to elicit robust humoral immunity to non-replicating antigens. We report that mice immunized with non-replicating Adjuplex-adjuvanted vaccines generated robust antigen-specific CTL responses. Vaccination by the subcutaneous or the intranasal route stimulated systemic and mucosal CTL memory respectively. However, only CTL memory induced by intranasal vaccination was protective against influenza viral challenge, and correlated with an enhancement of memory CTLs in the airways and CD103+ CD69+ CXCR3+ resident memory-like CTLs in the lungs. Mechanistically, Myd88-deficient mice mounted primary CTL responses to Adjuplex vaccines that were similar in magnitude to wild-type mice, but exhibited altered differentiation of effector cell subsets. Immune potentiating effects of Adjuplex entailed alterations in the frequency of antigen-presenting-cell subsets in vaccine draining lymph nodes, and in the lungs and airways following intranasal vaccination. Further, Adjuplex enhanced the ability of dendritic cells to promote antigen-induced proliferation of naïve CD8 T cells by modulating antigen uptake, its intracellular localization, and rate of processing. Taken together, we have identified an adjuvant that elicits both systemic and mucosal CTL memory to non-replicating antigens, and engenders protective CTL-based heterosubtypic immunity to influenza A virus in the respiratory tract. Further, findings presented in this manuscript have provided key insights into the mechanisms and factors that govern the induction and programming of systemic and protective memory CTLs in the respiratory tract. Current respiratory-virus vaccines typically employ non-replicating antigens and rely solely on the generation of humoral responses for protection. Viruses such as influenza can mutate and escape these responses, thereby limiting immunity and necessitating revaccination. Cell-mediated immunity (CMI) could provide broader protection by targeting viral components that infrequently mutate, however non-replicating vaccines capable of inducing CMI are not available. Impediments to vaccine development include an incomplete understanding of the nature of protective respiratory CMI and a lack of vaccine adjuvants capable of eliciting CMI to non-replicating antigens. Using a mouse model, we characterized the protective immunity afforded by CMI responses to non-replicating vaccines formulated with the adjuvant Adjuplex. We found that vaccination via either the subcutaneous or intranasal route was capable of inducing potent CMI responses. However, only intranasal vaccination protected against challenge with heterosubtypic influenza viruses. This protection correlated with enhancement of T cells with a resident-memory phenotype in the lungs. Additionally, mechanistic studies showed that Adjuplex affects antigen-presenting cells via activation and alteration of antigen uptake, processing, and presentation. The current studies: (1) identified an adjuvant that elicits protective CMI to respiratory viral pathogens; (2) suggested that stimulation of protective CMI in the respiratory tract requires intranasal vaccine delivery.
Collapse
Affiliation(s)
- David J Gasper
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America.,Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brandon Neldner
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin H Plisch
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hani Rustom
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Emily Carrow
- Advanced Bioadjuvants, Omaha, Nebraska, United States of America
| | - Hirotaka Imai
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - M Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
23
|
Chen Y, Deng X, Deng J, Zhou J, Ren Y, Liu S, Prusak DJ, Wood TG, Bao X. Functional motifs responsible for human metapneumovirus M2-2-mediated innate immune evasion. Virology 2016; 499:361-368. [PMID: 27743962 PMCID: PMC5102771 DOI: 10.1016/j.virol.2016.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 01/12/2023]
Abstract
Human metapneumovirus (hMPV) is a major cause of lower respiratory infection in young children. Repeated infections occur throughout life, but its immune evasion mechanisms are largely unknown. We recently found that hMPV M2-2 protein elicits immune evasion by targeting mitochondrial antiviral-signaling protein (MAVS), an antiviral signaling molecule. However, the molecular mechanisms underlying such inhibition are not known. Our mutagenesis studies revealed that PDZ-binding motifs, 29-DEMI-32 and 39-KEALSDGI-46, located in an immune inhibitory region of M2-2, are responsible for M2-2-mediated immune evasion. We also found both motifs prevent TRAF5 and TRAF6, the MAVS downstream adaptors, to be recruited to MAVS, while the motif 39-KEALSDGI-46 also blocks TRAF3 migrating to MAVS. In parallel, these TRAFs are important in activating transcription factors NF-kB and/or IRF-3 by hMPV. Our findings collectively demonstrate that M2-2 uses its PDZ motifs to launch the hMPV immune evasion through blocking the interaction of MAVS and its downstream TRAFs.
This manuscript describes a molecular mechanism underlying the immune evasion of hMPV. Results create the design basis for safer and more effective hMPV vaccines/therapeutic molecules. We demonstrate the contribution of TRAFs in antiviral responses to hMPV infection.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pediatrics, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, China; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Xiaoling Deng
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Junfang Deng
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States; Department of Hepatobiliary Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Jiehua Zhou
- Department of Pediatrics, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, China
| | - Yuping Ren
- Department of Pediatrics, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, China; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Shengxuan Liu
- Department of Pediatrics, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, China; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Deborah J Prusak
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Thomas G Wood
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Xiaoyong Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, United States; The Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, United States; The Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
24
|
Li X, Guo L, Kong M, Su X, Yang D, Zou M, Liu Y, Lu L. Design and Evaluation of a Multi-Epitope Peptide of Human Metapneumovirus. Intervirology 2016; 58:403-12. [PMID: 27096202 DOI: 10.1159/000445059] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/23/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES No licensed vaccines or therapeutic agents for human metapneumovirus (hMPV) infection exist to date. We aimed to construct a multi-epitope peptide (MEP) of hMPV to show promising results for epitope-based vaccine development. METHODS Six independent algorithms were screened to predict B-cell epitopes of hMPV, and three algorithms were used to predict cytotoxic T lymphocyte and T helper (Th) lymphocyte epitopes. Predicted epitopes were assembled in series with the spacers GPGPG and KK introduced, termed MEP. Recombinant mep genes were inserted into pET32a(+) plasmid and expressed in Escherichia coli strain BL21 (DE3). BALB/c mice were immunized with MEP with different adjuvants. Antibody titer, lymphocyte proliferation, cytotoxic T lymphocyte (CTL) activity and splenocyte cytokines were detected 2 weeks later after the last immunization. Microneutralization assay was used to detect neutralizing antibodies. RESULTS Six B-cell epitopes, four CTL epitopes and two Th epitopes were screened to construct the mep gene. Expressed MEP induced >104 antibodies in BALB/c mice, and produced anti-MEP antibody reacting with hMPV strains specifically as detected in indirect fluorescent assay (the titer was 160). The lymphocyte proliferation index, CTL activity and splenocyte cytokines of the MEP immunization groups were higher than in the control group (p < 0.05). Both IgG1 and IgG2a antibodies could be detected in the different groups, and balanced Th1/Th2 cytokines were secreted by splenocytes in these groups. The mean neutralizing titers of the MEP+CpG ODN, MEP+Alum and MEP+Alum+ CpG ODN groups were 87 (95% CI 50-126), 93 (95% CI 67-121) and 96 (95% CI 69-147), respectively. CONCLUSION MEP of hMPV elicited both strong humoral immunity and cell-mediated immunity in mice. The anti-MEP serum could neutralize hMPV infection in vitro. Joint use of CpG ODN and aluminum hydroxide adjuvants obtained the best immune effects. This study may contribute to hMPV epitope-based vaccine development.
Collapse
Affiliation(s)
- Xiaoyan Li
- Tianjin Centers for Disease Control and Prevention, Tianjin, PR China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Hastings AK, Gilchuk P, Joyce S, Williams JV. Novel HLA-A2-restricted human metapneumovirus epitopes reduce viral titers in mice and are recognized by human T cells. Vaccine 2016; 34:2663-70. [PMID: 27105560 DOI: 10.1016/j.vaccine.2016.04.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 11/25/2022]
Abstract
Human metapneumovirus (HMPV) is a major cause of morbidity and mortality from acute lower respiratory tract illness, with most individuals seropositive by age five. Despite the presence of neutralizing antibodies, secondary infections are common and can be severe in young, elderly, and immunocompromised persons. Preclinical vaccine studies for HMPV have suggested a need for a balanced antibody and T cell immune response to enhance protection and avoid lung immunopathology. We infected transgenic mice expressing human HLA-A*0201 with HMPV and used ELISPOT to screen overlapping and predicted epitope peptides. We identified six novel HLA-A2 restricted CD8(+) T cell (TCD8) epitopes, with M39-47 (M39) immunodominant. Tetramer staining detected M39-specific TCD8 in lungs and spleen of HMPV-immune mice. Immunization with adjuvant-formulated M39 peptide reduced lung virus titers upon challenge. Finally, we show that TCD8 from HLA-A*0201 positive humans recognize M39 by IFNγ ELISPOT and tetramer staining. These results will facilitate HMPV vaccine development and human studies.
Collapse
Affiliation(s)
- Andrew K Hastings
- Department of Pathology, Microbiology & Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology & Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology & Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Veterans Administration Tennessee Valley Healthcare System, Nashville, TN 37332, USA
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, USA.
| |
Collapse
|
26
|
Immune Response to Human Metapneumovirus Infection: What We Have Learned from the Mouse Model. Pathogens 2015; 4:682-96. [PMID: 26393657 PMCID: PMC4584281 DOI: 10.3390/pathogens4030682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023] Open
Abstract
Human Metapneumovirus (hMPV) is a leading respiratory viral pathogen associated with bronchiolitis, pneumonia, and asthma exacerbation in young children, the elderly and immunocompromised individuals. The development of a potential vaccine against hMPV requires detailed understanding of the host immune system, which plays a significant role in hMPV pathogenesis, susceptibility and vaccine efficacy. As a result, animal models have been developed to better understand the mechanisms by which hMPV causes disease. Several animal models have been evaluated and established so far to study the host immune responses and pathophysiology of hMPV infection. However, inbred laboratory mouse strains have been one of the most used animal species for experimental modeling and therefore used for the studies of immunity and immunopathogenesis to hMPV. This review summarizes the contributions of the mouse model to our understanding of the immune response against hMPV infection.
Collapse
|
27
|
New Approaches for Immunization and Therapy against Human Metapneumovirus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:858-66. [PMID: 26063237 DOI: 10.1128/cvi.00230-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Human metapneumovirus (HMPV) is a paramyxovirus discovered in 2001 in the Netherlands. Studies have identified HMPV as an important causative agent of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. There are currently no licensed therapeutics or vaccines against HMPV. However, several research groups have tested vaccine candidates and monoclonal antibodies in various animal models. Several of these approaches have shown promise in animal models. This minireview summarizes the current therapies used to treat HMPV infection as well as different approaches for immunization.
Collapse
|
28
|
Lung CD8+ T Cell Impairment Occurs during Human Metapneumovirus Infection despite Virus-Like Particle Induction of Functional CD8+ T Cells. J Virol 2015; 89:8713-26. [PMID: 26063431 DOI: 10.1128/jvi.00670-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/04/2015] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a major cause of respiratory disease in infants, the elderly, and immunocompromised individuals worldwide. There is currently no licensed HMPV vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate because they are noninfectious and elicit a neutralizing antibody response. However, studies show that serum neutralizing antibodies are insufficient for complete protection against reinfection and that adaptive T cell immunity is important for viral clearance. HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment, mediated by programmed death 1 (PD-1). In this study, we generated HMPV VLPs by expressing the fusion and matrix proteins in mammalian cells and tested whether VLP immunization induces functional HMPV-specific TCD8 responses in mice. C57BL/6 mice vaccinated twice with VLPs and subsequently challenged with HMPV were protected from lung viral replication for at least 20 weeks postimmunization. A single VLP dose elicited F- and M-specific lung TCD8s with higher function and lower expression of PD-1 and other inhibitory receptors than TCD8s from HMPV-infected mice. However, after HMPV challenge, lung TCD8s from VLP-vaccinated mice exhibited inhibitory receptor expression and functional impairment similar to those of mice experiencing secondary infection. HMPV challenge of VLP-immunized μMT mice also elicited a large percentage of impaired lung TCD8s, similar to mice experiencing secondary infection. Together, these results indicate that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of acute respiratory disease for which there is no licensed vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate and induce antibodies, but T cell responses are less defined. Moreover, HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment mediated by programmed death 1 (PD-1). In this study, HMPV VLPs containing viral fusion and matrix proteins elicited epitope-specific TCD8s that were functional with low PD-1 expression. Two VLP doses conferred sterilizing immunity in C57BL/6 mice and facilitated HMPV clearance in antibody-deficient μMT mice without enhancing lung pathology. However, regardless of whether responding lung TCD8s had previously encountered HMPV antigens in the context of VLPs or virus, similar proportions were impaired and expressed comparable levels of PD-1 upon viral challenge. These results suggest that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge.
Collapse
|
29
|
Deng J, Chen Y, Liu G, Ren J, Go C, Ivanciuc T, Deepthi K, Casola A, Garofalo RP, Bao X. Mitochondrial antiviral-signalling protein plays an essential role in host immunity against human metapneumovirus. J Gen Virol 2015; 96:2104-2113. [PMID: 25953917 DOI: 10.1099/vir.0.000178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human metapneumovirus (hMPV) is a common cause of respiratory tract infection in the paediatrics population. Recently, we and others have shown that retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs) are essential for hMPV-induced cellular antiviral signalling. However, the contribution of those receptors to host immunity against pulmonary hMPV infection is largely unexplored. In this study, mice deficient in mitochondrial antiviral-signalling protein (MAVS), an adaptor of RLRs, were used to investigate the role(s) of these receptors in pulmonary immune responses to hMPV infection. MAVS deletion significantly impaired the induction of antiviral and pro-inflammatory cytokines and the recruitment of immune cells to the bronchoalveolar lavage fluid by hMPV. Compared with WT mice, mice lacking MAVS demonstrated decreased abilities to activate pulmonary dendritic cells (DCs) and abnormal primary T-cell responses to hMPV infection. In addition, mice deficient in MAVS had a higher peak of viral load at day 5 post-infection (p.i.) than WT mice, but were able to clear hMPV by day 7 p.i. similarly to WT mice. Taken together, our data indicate a role of MAVS-mediated pathways in the pulmonary immune responses to hMPV infection and the early control of hMPV replication.
Collapse
Affiliation(s)
- Junfang Deng
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Hepatobiliary Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, PR China
| | - Yu Chen
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pediatrics, TongJi Hospital, Huazhong University of Science and Technology, PR China
| | - Guangliang Liu
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Otorhinolaryngology, Sixth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Junping Ren
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Caroline Go
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Kolli Deepthi
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Roberto P Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaoyong Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
30
|
Abstract
Human metapneumovirus (hMPV) and respiratory syncytial virus, its close family member, are two major causes of lower respiratory tract infection in the paediatric population. hMPV is also a common cause of worldwide morbidity and mortality in immunocompromised patients and older adults. Repeated infections occur often, demonstrating a heavy medical burden. However, there is currently no hMPV-specific prevention treatment. This review focuses on the current literature on hMPV vaccine development. We believe that a better understanding of the role(s) of viral proteins in host responses might lead to efficient prophylactic vaccine development.
Collapse
Affiliation(s)
- J Ren
- 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - T Phan
- 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - X Bao
- 2Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA 3Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
31
|
Acute clearance of human metapneumovirus occurs independently of natural killer cells. J Virol 2014; 88:10963-9. [PMID: 24965465 DOI: 10.1128/jvi.01558-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory disease. The role of NK cells in protection against HMPV is unclear. We show that while HMPV-infected C57BL/6 mice had higher numbers of functional lung NK cells than mock-treated mice, comparing NK cell-depleted and control mice did not reveal differences in lung viral titers, histopathology, cytokine levels, or T cell numbers or function. These data indicate that NK cells are not required for host control of HMPV.
Collapse
|
32
|
Panda S, Mohakud NK, Pena L, Kumar S. Human metapneumovirus: review of an important respiratory pathogen. Int J Infect Dis 2014; 25:45-52. [PMID: 24841931 PMCID: PMC7110553 DOI: 10.1016/j.ijid.2014.03.1394] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/18/2014] [Accepted: 03/27/2014] [Indexed: 12/31/2022] Open
Abstract
Human metapneumovirus (hMPV), discovered in 2001, most commonly causes upper and lower respiratory tract infections in young children, but is also a concern for elderly subjects and immune-compromised patients. hMPV is the major etiological agent responsible for about 5% to 10% of hospitalizations of children suffering from acute respiratory tract infections. hMPV infection can cause severe bronchiolitis and pneumonia in children, and its symptoms are indistinguishable from those caused by human respiratory syncytial virus. Initial infection with hMPV usually occurs during early childhood, but re-infections are common throughout life. Due to the slow growth of the virus in cell culture, molecular methods (such as reverse transcriptase PCR (RT-PCR)) are the preferred diagnostic modality for detecting hMPV. A few vaccine candidates have been shown to be effective in preventing clinical disease, but none are yet commercially available. Our understanding of hMPV has undergone major changes in recent years and in this article we will review the currently available information on the molecular biology and epidemiology of hMPV. We will also review the current therapeutic interventions and strategies being used to control hMPV infection, with an emphasis on possible approaches that could be used to develop an effective vaccine against hMPV.
Collapse
Affiliation(s)
- Swagatika Panda
- School of Biotechnology, KIIT University, Campus XI, Patia, Bhubaneswar 751024, Orissa, India
| | - Nirmal Kumar Mohakud
- Department of Paediatrics, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Orissa, India
| | - Lindomar Pena
- Department of Cell and Molecular Biology, Centre for Biotechnology, Federal University of Paraiba, Joao Pessoa, Paraiba, Brazil
| | - Subrat Kumar
- School of Biotechnology, KIIT University, Campus XI, Patia, Bhubaneswar 751024, Orissa, India.
| |
Collapse
|
33
|
Human metapneumovirus virus-like particles induce protective B and T cell responses in a mouse model. J Virol 2014; 88:6368-79. [PMID: 24672031 DOI: 10.1128/jvi.00332-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a leading cause of respiratory disease in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach because of reduced safety concerns compared with live vaccines. We generated HMPV VLPs by expressing viral proteins in suspension-adapted human embryonic kidney epithelial (293-F) cells and found that the viral matrix (M) and fusion (F) proteins were sufficient to form VLPs. We previously reported that the VLPs resemble virus morphology and incorporate fusion-competent F protein (R. G. Cox, S. B. Livesay, M. Johnson, M. D. Ohi, and J. V. Williams, J. Virol. 86:12148-12160, 2012), which we hypothesized would elicit F-specific antibody and T cell responses. In this study, we tested whether VLP immunization could induce protective immunity to HMPV by using a mouse model. C57BL/6 mice were injected twice intraperitoneally with VLPs alone or with adjuvant and subsequently challenged with HMPV. Mice were euthanized 5 days postinfection, and virus titers, levels of neutralizing antibodies, and numbers of CD3(+) T cells were quantified. Mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant, though adjuvant reduced cellular immune responses. Two doses of VLPs conferred complete protection from HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach. We generated HMPV VLPs by expressing the viral matrix (M) and fusion (F) proteins in mammalian cells. We found that mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant. Two doses of VLPs conferred complete protection against HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV.
Collapse
|
34
|
Medina-Armenteros Y, Farinha-Arcieri LE, Braga CJM, Carromeu C, Tamura RE, Ventura AM. Mapping of CD8 T cell epitopes in human respiratory syncytial virus L protein. Intervirology 2014; 57:55-64. [PMID: 24480928 DOI: 10.1159/000357325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 11/07/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Since it has been reported that in humans there is a relationship between human respiratory syncytial virus (hRSV)-specific cytotoxic T lymphocytes and symptom reduction, and that the polymerase (structural L protein) is highly conserved among different strains, this work aimed to identify the CD8 T cell epitopes H-2(d) restricted within the L sequence for immunization purposes. METHODS We screened the hRSV strain A2 L protein sequence using two independent algorithms, SYFPEITHI and PRED/(BALB/c), to predict CD8 T cell epitopes. The selected peptides were synthesized and used to immunize BALB/c mice for the evaluation of T cell response. The production of IFN-γ from splenocytes of hRSV-infected animals stimulated by these peptides was assayed by ELISPOT. RESULTS Nine peptides showing the best binding scores to the BALB/c MHC-I molecules (H-2K(d), L(d) and D(d)) were selected. Sequence homology analysis showed that these sequences are conserved among different hRSV strains. Two of these peptides induced significant IFN-γ production by ex vivo-stimulated T cells. CONCLUSIONS Our results indicate that the hRSV L protein contains H-2(d)-restricted epitopes.
Collapse
Affiliation(s)
- Yordanka Medina-Armenteros
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
35
|
Liu P, Shu Z, Qin X, Dou Y, Zhao Y, Zhao X. A live attenuated human metapneumovirus vaccine strain provides complete protection against homologous viral infection and cross-protection against heterologous viral infection in BALB/c mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1246-1254. [PMID: 23761661 PMCID: PMC3754520 DOI: 10.1128/cvi.00145-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/05/2013] [Indexed: 11/20/2022]
Abstract
A live attenuated vaccine candidate strain (M2) of human metapneumovirus (hMPV) was generated by removing the N-linked carbohydrate at amino acid 172 in the fusion (F) protein. Previously, replication of M2 in mouse lungs could be detected by molecular assays but not by viral titration. In the present study, the protective effects of M2 against infection by homologous or heterologous viruses were evaluated in BALB/c mice. Immunization with M2 produced a high titer of serum virus-neutralizing antibodies in BALB/c mice at 4 and 8 weeks postimmunization, with the titers against the homologous virus being higher than those against the heterologous virus. Challenges at 4 and 8 weeks postinoculation with M2 or wild-type virus led to no replication when mice were challenged with a homologous virus and extremely reduced replication when mice were challenged with a heterologous virus, as determined by the detection of viral genomic RNA copies in the lungs, as well as significantly milder pulmonary pathology. Thus, M2, with only one N-linked carbohydrate removed in the F protein, provides complete protection from homologous virus infection and substantial cross-protection from heterologous virus infection for at least 56 days after inoculation. This vaccine strain may therefore be a candidate for further preclinical study. Furthermore, this attenuating strategy (changing the glycosylation of a major viral protein) may be useful in the development of other viral vaccines.
Collapse
Affiliation(s)
- Ping Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
36
|
Ren J, Kolli D, Deng J, Fang R, Gong B, Xue M, Casola A, Garofalo RP, Wang T, Bao X. MyD88 controls human metapneumovirus-induced pulmonary immune responses and disease pathogenesis. Virus Res 2013; 176:241-50. [PMID: 23845303 DOI: 10.1016/j.virusres.2013.06.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/23/2013] [Accepted: 06/26/2013] [Indexed: 12/14/2022]
Abstract
Human metapneumovirus (hMPV) is a common cause of lung and airway infections in infants and young children. Recently, we and others have shown that hMPV infection induces Toll-like receptor (TLR)-dependent cellular signaling. However, the contribution of TLR-mediated signaling in host defenses against pulmonary hMPV infection and associated disease pathogenesis has not been elucidated. In this study, mice deficient in MyD88, a common adaptor of TLRs, was used to investigate the contribution of TLRs to in vivo pulmonary response to hMPV infection. MyD88(-/-) mice have significantly reduced pulmonary inflammation and associated disease compared with wild-type (WT) C57BL/6 mice after intranasal infection with hMPV. hMPV-induced cytokines and chemokines in bronchoalveolar lavage fluid (BALF) and isolated lung conventional dendritic cells (cDC) are also significantly impaired by MyD88 deletion. In addition, we found that MyD88 is required for the recruitment of DC, T cells, and other immune cells to the lungs, and for the functional regulation of DC and T cells in response to hMPV infection. Taken together, our data indicate that MyD88-mediated pathways are essential for the pulmonary immune and pathogenic responses to this viral pathogen.
Collapse
Affiliation(s)
- Junping Ren
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yang CF, Wang CK, Tollefson SJ, Lintao LD, Liem A, Chu M, Williams JV. Human metapneumovirus G protein is highly conserved within but not between genetic lineages. Arch Virol 2013; 158:1245-52. [PMID: 23385328 DOI: 10.1007/s00705-013-1622-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/17/2012] [Indexed: 11/30/2022]
Abstract
Human metapneumovirus (HMPV) is an important cause of acute respiratory illnesses in children. HMPV encodes two major surface glycoproteins, fusion (F) and glycoprotein (G). The function of G has not been fully established, though it is dispensable for in vitro and in vivo replication. We analyzed 87 full-length HMPV G sequences from isolates collected over 20 years. The G sequences fell into four subgroups with a mean 63 % amino acid identity (minimum 29 %). The length of G varied from 217 to 241 residues. Structural features such as proline content and N- and O-glycosylation sites were present in all strains but quite variable between subgroups. There was minimal drift within the subgroups over 20 years. The estimated time to the most recent common ancestor was 215 years. HMPV G was conserved within lineages over 20 years, suggesting functional constraints on diversity. However, G was poorly conserved between subgroups, pointing to potentially distinct roles for G among different viral lineages.
Collapse
|
38
|
Abstract
It has been 10 years since human metapneumovirus (HMPV) was identified as a causative agent of respiratory illness in humans. Since then, numerous studies have contributed to a substantial body of knowledge on many aspects of HMPV. This review summarizes our current knowledge on HMPV, HMPV disease pathogenesis, and disease intervention strategies and identifies a number of areas with key questions to be addressed in the future.
Collapse
|
39
|
Zhao K, Wang H, Wu C. The immune responses of HLA-A*0201 restricted SARS-CoV S peptide-specific CD8⁺ T cells are augmented in varying degrees by CpG ODN, PolyI:C and R848. Vaccine 2011; 29:6670-8. [PMID: 21745520 PMCID: PMC7115397 DOI: 10.1016/j.vaccine.2011.06.100] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 06/13/2011] [Accepted: 06/25/2011] [Indexed: 01/13/2023]
Abstract
The induction of antigen specific memory CD8+ T cells in vivo is very important to new vaccines against infectious diseases. In the present study, we aimed to evaluate the immune responses of peptide-specific CD8+ T cells induced by HLA-A*0201 restricted severe acute respiratory syndrome-associated coronavirus (SARS-CoV) S epitopes plus CpG oligodeoxynucleotide (CpG ODN), PolyI:C and R848 as adjuvants. Furthermore, the generation, distribution and phenotype of long-lasting peptide-specific memory CD8+ T cells were assessed by ELISA, ELISPOT and flow cytometry. Our results showed that antigen specific CD8+ T cells were elicited by HLA-A*0201 restricted SARS-CoV S epitopes. Furthermore, the frequency of peptide-specific CD8+ T cells was dramatically increased after both prime and boost immunization with peptides plus CpG ODN, whereas slight enhancements were induced following boost vaccination with peptides plus PolyI:C or R848. SARS-CoV S peptide-specific IFN-γ+CD8+ T cells were distributed throughout the lymphoid and non-lymphoid tissues. Results also demonstrated that the HLA-A*0201 restricted peptide-specific CD8+ T cells induced by peptides plus CpG ODN carried a memory cell phenotype with CD45RB+ and CD62L− and possessed long-term survival ability in vivo. Taken together, our results implied that HLA-A*0201 restricted SARS-CoV S epitopes plus CpG ODN might be the superior candidates for SARS vaccine.
Collapse
Affiliation(s)
- Kai Zhao
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province 510080, People' s Republic of China
| | | | | |
Collapse
|
40
|
Gaunt ER, Jansen RR, Poovorawan Y, Templeton KE, Toms GL, Simmonds P. Molecular epidemiology and evolution of human respiratory syncytial virus and human metapneumovirus. PLoS One 2011; 6:e17427. [PMID: 21390255 PMCID: PMC3046979 DOI: 10.1371/journal.pone.0017427] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 02/02/2011] [Indexed: 11/23/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV) are ubiquitous respiratory pathogens of the Pneumovirinae subfamily of the Paramyxoviridae. Two major surface antigens are expressed by both viruses; the highly conserved fusion (F) protein, and the extremely diverse attachment (G) glycoprotein. Both viruses comprise two genetic groups, A and B. Circulation frequencies of the two genetic groups fluctuate for both viruses, giving rise to frequently observed switching of the predominantly circulating group. Nucleotide sequence data for the F and G gene regions of HRSV and HMPV variants from the UK, The Netherlands, Bangkok and data available from Genbank were used to identify clades of both viruses. Several contemporary circulating clades of HRSV and HMPV were identified by phylogenetic reconstructions. The molecular epidemiology and evolutionary dynamics of clades were modelled in parallel. Times of origin were determined and positively selected sites were identified. Sustained circulation of contemporary clades of both viruses for decades and their global dissemination demonstrated that switching of the predominant genetic group did not arise through the emergence of novel lineages each respiratory season, but through the fluctuating circulation frequencies of pre-existing lineages which undergo proliferative and eclipse phases. An abundance of sites were identified as positively selected within the G protein but not the F protein of both viruses. For HRSV, these were discordant with previously identified residues under selection, suggesting the virus can evade immune responses by generating diversity at multiple sites within linear epitopes. For both viruses, different sites were identified as positively selected between genetic groups.
Collapse
Affiliation(s)
- Eleanor R Gaunt
- Centre for Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | |
Collapse
|
41
|
Chen P, Rayner S, Hu KH. Advances of bioinformatics tools applied in virus epitopes prediction. Virol Sin 2011; 26:1-7. [PMID: 21331885 PMCID: PMC7090880 DOI: 10.1007/s12250-011-3159-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 10/16/2010] [Indexed: 11/18/2022] Open
Abstract
In recent years, the in silico epitopes prediction tools have facilitated the progress of vaccines development significantly and many have been applied to predict epitopes in viruses successfully. Herein, a general overview of different tools currently available, including T cell and B cell epitopes prediction tools, is presented. And the principles of different prediction algorithms are reviewed briefly. Finally, several examples are present to illustrate the application of the prediction tools.
Collapse
Affiliation(s)
- Ping Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | | | | |
Collapse
|
42
|
CD8+ T cell response in HLA-A*0201 transgenic mice is elicited by epitopes from SARS-CoV S protein. Vaccine 2010; 28:6666-74. [PMID: 20709007 PMCID: PMC7115361 DOI: 10.1016/j.vaccine.2010.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 07/06/2010] [Accepted: 08/02/2010] [Indexed: 11/20/2022]
Abstract
Cytotoxic CD8(+) T lymphocytes (CTLs) play an important role in antiviral immunity. Several human HLA-A*0201 restricted CTL epitopes of severe acute respiratory syndrome (SARS) spike (S) protein have been identified in HLA-A*0201 transgenic (Tg) mice, but the mechanisms and properties of immune responses are still not well understood. In this study, HLA-A*0201 Tg mice were primed intramuscularly with SARS S DNA and boosted subcutaneously with HLA-A*0201 restricted peptides. The lymphocytes from draining lymph nodes, spleens and lungs were stimulated with the cognate peptides. Three different methods (ELISA, ELISPOT and FACS) were used to evaluate the immune responses during short and long periods of time after immunization. Results showed that peptide-specific CD8(+) T cells secreted IFN-γ, TNF-α and IL-2 and expressed CD107a/b on cell surface. IFN-γ(+)CD8(+) T cells and CD107a/b(+)CD8(+) T cells distributed throughout the lymphoid and non-lymphoid tissues, but the frequency of peptide-specific CD8(+) T cells was higher in lungs than in spleens and lymph nodes. The phenotype of the CD8(+) T cells was characterized based on the expression of IFN-γ. Most of the HLA-A*0201 restricted peptide-specific CD8(+) T cells represented a memory subset with CD45RB(high) and CD62L(low). Taken together, these data demonstrate that immunization with SARS S DNA and HLA-A*0201 restricted peptides can elicit antigen-specific CD8(+) T cell immune responses which may have a significant implication in the long-term protection. We provide novel information in cellular immune responses of SARS S antigen-specific CD8(+) T cells, which are important in the development of vaccine against SARS-CoV infection.
Collapse
|
43
|
Ryder AB, Tollefson SJ, Podsiad AB, Johnson JE, Williams JV. Soluble recombinant human metapneumovirus G protein is immunogenic but not protective. Vaccine 2010; 28:4145-52. [PMID: 20417260 PMCID: PMC2894472 DOI: 10.1016/j.vaccine.2010.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 12/17/2009] [Accepted: 04/07/2010] [Indexed: 11/28/2022]
Abstract
Human metapneumovirus (HMPV) expresses the major surface glycoproteins F and G. We evaluated the protective efficacy of immunization with G. We generated a recombinant form of G ectodomain (GDeltaTM) that was secreted from mammalian cells and purified by affinity chromatography. We tested the immunogenicity of GDeltaTM in cotton rats. Animals were immunized with PBS, GDeltaTM alone or adjuvanted, or were infected once with HMPV, and challenged with live HMPV at 28 days. Animals vaccinated with adjuvanted and non-adjuvanted GDeltaTM developed high levels of serum antibodies to both recombinant and native G protein; however, vaccinated animals did not develop neutralizing antibodies and were not protected against virus challenge. Unlike the analogous non-fusion glycoproteins of other human paramyxoviruses, HMPV G does not appear to be a protective antigen. This represents an unusual feature of HMPV.
Collapse
Affiliation(s)
- Alex B. Ryder
- Vanderbilt University Medical Center, School of Medicine, Nashville, TN, USA
| | | | | | | | - John V. Williams
- Department of Pediatrics, Nashville, TN, USA
- Department of Microbiology and Immunology, Nashville, TN, USA
| |
Collapse
|
44
|
Chen D, Edgtton K, Gould A, Guo H, Mather M, Haigh O, Cochrane M, Kattenbelt J, Thomson S, Tindle R. HBsAg-vectored vaccines simultaneously deliver CTL responses to protective epitopes from multiple viral pathogens. Virology 2010; 398:68-78. [DOI: 10.1016/j.virol.2009.11.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2009] [Accepted: 11/24/2009] [Indexed: 01/12/2023]
|
45
|
Herd KA, Nelson M, Mahalingam S, Tindle RW. Pulmonary infection of mice with human metapneumovirus induces local cytotoxic T-cell and immunoregulatory cytokine responses similar to those seen with human respiratory syncytial virus. J Gen Virol 2010; 91:1302-10. [PMID: 20053825 DOI: 10.1099/vir.0.015396-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human metapneumovirus (hMPV) is a major cause of upper and lower respiratory-tract infection in infants, the elderly and immunocompromised individuals. Virus-directed cellular immunity elicited by hMPV infection is poorly understood, in contrast to the phylogenetically and clinically related pathogen human respiratory syncytial virus (hRSV). In a murine model of acute lower respiratory-tract infection with hMPV, we demonstrate the accumulation of gamma interferon (IFN-gamma)-producing CD8+ T cells in the airways and lungs at day 7 post-infection (p.i.), associated with cytotoxic T lymphocytes (CTLs) directed to an epitope of the M2-1 protein. This CTL immunity was accompanied by increased pulmonary expression of Th1 cytokines IFN-gamma and interleukin (IL)-12 and antiviral cytokines (IFN-beta), as well as chemokines Mip-1alpha, Mip-1beta, Mig, IP-10 and CX3CL1. There was also a moderate increase in Th2-type cytokines IL-4 and IL-10 compared with uninfected mice. At 21 days p.i., a strong CTL response could be recalled from the spleen. A similar pattern of CTL induction to the homologous M2-1 CTL epitope of hRSV, and of cytokine/chemokine induction, was observed following infection with hRSV, highlighting similarities in the cellular immune response to the two related pathogens.
Collapse
Affiliation(s)
- Karen A Herd
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, and Clinical Medical Virology Centre, University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
46
|
Lim HJ, Lee JW, Park YS, Kim NH, Kim M, Yim JJ, Yang SC, Yoo CG, Kim YW, Han SK, Shim YS, Lee SM. A Case of Severe Human Metapneumovirus Pneumonia Requiring Mechanical Ventilation in an Immunocompetent Adult. Tuberc Respir Dis (Seoul) 2009. [DOI: 10.4046/trd.2009.67.2.135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hyo-Jeong Lim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Woo Lee
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young Sik Park
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Nak-Hyun Kim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Moonsuk Kim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Joon Yim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seok-Chul Yang
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Gyu Yoo
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young Whan Kim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Koo Han
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Soo Shim
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Min Lee
- Department of Internal Medicine and Lung Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Mok H, Tollefson SJ, Podsiad AB, Shepherd BE, Polosukhin VV, Johnston RE, Williams JV, Crowe JE. An alphavirus replicon-based human metapneumovirus vaccine is immunogenic and protective in mice and cotton rats. J Virol 2008; 82:11410-8. [PMID: 18786987 PMCID: PMC2573258 DOI: 10.1128/jvi.01688-08] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 09/02/2008] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (hMPV) is a recently discovered paramyxovirus that causes upper and lower respiratory tract infections in infants, the elderly, and immunocompromised individuals worldwide. Here, we developed Venezuelan equine encephalitis virus replicon particles (VRPs) encoding hMPV fusion (F) or attachment (G) glycoproteins and evaluated the immunogenicity and protective efficacy of these vaccine candidates in mice and cotton rats. VRPs encoding hMPV F protein, when administered intranasally, induced F-specific virus-neutralizing antibodies in serum and immunoglobulin A (IgA) antibodies in secretions at the respiratory mucosa. Challenge virus replication was reduced significantly in both the upper and lower respiratory tracts following intranasal hMPV challenge in these animals. However, vaccination with hMPV G protein VRPs did not induce neutralizing antibodies or protect animals from hMPV challenge. Close examination of the histopathology of the lungs of VRP-MPV F-vaccinated animals following hMPV challenge revealed no enhancement of inflammation or mucus production. Aberrant cytokine gene expression was not detected in these animals. Together, these results represent an important first step toward the use of VRPs encoding hMPV F proteins as a prophylactic vaccine for hMPV.
Collapse
Affiliation(s)
- Hoyin Mok
- Departments of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lidbury BA, Rulli NE, Suhrbier A, Smith PN, McColl SR, Cunningham AL, Tarkowski A, van Rooijen N, Fraser RJ, Mahalingam S. Macrophage-derived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J Infect Dis 2008; 197:1585-93. [PMID: 18433328 DOI: 10.1086/587841] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Alphaviruses, such as chikungunya virus and Ross River virus (RRV), are associated with outbreaks of infectious rheumatic disease in humans worldwide. Using an established mouse model of disease that mimics RRV disease in humans, we showed that macrophage-derived factors are critical in the development of striated muscle and joint tissue damage. Histologic analyses of muscle and ankle joint tissues demonstrated a substantial reduction in inflammatory infiltrates in infected mice depleted of macrophages (i.e., "macrophage-depleted mice"). Levels of the proinflammatory factors tumor necrosis factor-alpha, interferon-gamma, and macrophage chemoattractant protein-1 were also dramatically reduced in tissue samples obtained from infected macrophage-depleted mice, compared with samples obtained from infected mice without macrophage depletion. These factors were also detected in the synovial fluid of patients with RRV-induced polyarthritis. Neutralization of these factors reduced the severity of disease in mice, whereas blocking nuclear factor kappaB by treatment with sulfasalazine ameliorated RRV inflammatory disease and tissue damage. To our knowledge, these findings are the first to demonstrate that macrophage-derived products play important roles in the development of arthritis and myositis triggered by alphavirus infection.
Collapse
Affiliation(s)
- Brett A Lidbury
- Virus and Inflammation Research Group, Faculty of Sciences, University of Canberra, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tauro S, Su YC, Thomas S, Schwarze J, Matthaei KI, Townsend D, Simson L, Tripp RA, Mahalingam S. Molecular and cellular mechanisms in the viral exacerbation of asthma. Microbes Infect 2008; 10:1014-23. [PMID: 18762266 PMCID: PMC7172315 DOI: 10.1016/j.micinf.2008.07.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The aetiology of asthma associated with viral infection is complex. The dynamics that contribute to disease pathogenesis are multifactorial and involve overlapping molecular and cellular mechanisms, particularly the immune response to respiratory virus infection or allergen sensitization. This review summarizes the evidence associated with factors that may contribute to the development or exacerbation of asthma including age, host factors, genetic polymorphisms, altered immune responses, and aspects of viral antigen expression. This review also provides an important perspective of key events linked to the development of asthmatic disease and related pulmonary inflammation from human and animal studies, and discusses their relationship as targets for disease intervention strategies.
Collapse
Affiliation(s)
- Sharyn Tauro
- Centre for Biomolecular and Chemical Sciences, University of Canberra, Canberra, ACT 2601, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Herfst S, de Graaf M, Schrauwen EJA, Sprong L, Hussain K, van den Hoogen BG, Osterhaus ADME, Fouchier RAM. Generation of temperature-sensitive human metapneumovirus strains that provide protective immunity in hamsters. J Gen Virol 2008; 89:1553-1562. [PMID: 18559924 DOI: 10.1099/vir.0.2008/002022-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human metapneumovirus (HMPV) causes acute respiratory tract illness primarily in young children, immunocompromised individuals and the elderly. Vaccines would be desirable to prevent severe illnesses in these risk groups. Here, we describe the generation and evaluation of cold-passage (cp) temperature-sensitive (ts) HMPV strains as vaccine candidates. Repeated passage of HMPV at low temperatures in Vero cells resulted in the accumulation of mutations in the viral genome. Introduction of these mutations in a recombinant HMPV by reverse genetics resulted in a ts-phenotype, judged on the decreased shut-off temperature for virus replication in vitro. As an alternative approach, three previously described cp-respiratory syncytial virus (cp-HRSV) mutations were introduced in a recombinant HMPV, which also resulted in a low shut-off temperature in vitro. Replication of these ts-viruses containing either the cp-HMPV or cp-HRSV mutations was reduced in the upper respiratory tract (URT) and undetectable in the lower respiratory tract (LRT) of hamsters. Nevertheless, high titres of HMPV-specific antibodies were induced by both ts-viruses. Upon immunization with the ts-viruses, the LRT of hamsters were completely protected against challenge infection with a heterologous HMPV strain, and URT viral titres were reduced by 10 000-fold. In conclusion, we provide proof-of-principle for two candidate live-attenuated HMPV vaccines that induce cross-protective immunity to prevent infection of the LRT in Syrian golden hamsters. Further mapping of the molecular determinants of attenuation of HMPV should be the subject of future studies.
Collapse
Affiliation(s)
- Sander Herfst
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Miranda de Graaf
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Leo Sprong
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Karim Hussain
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Ron A M Fouchier
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|