1
|
Bajinka O, Ouedraogo SY, Li N, Zhan X. Multiomics as instrument to promote 3P medical approaches for the overall management of respiratory syncytial viral infections. EPMA J 2025; 16:217-238. [PMID: 39991100 PMCID: PMC11842696 DOI: 10.1007/s13167-024-00395-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/29/2024] [Indexed: 02/25/2025]
Abstract
Respiratory syncytial viral (RSV) infection is a leading persisting pulmonary disease-causing agent. It causes loss of lives especially among infants, old ages, and adults immunocompromised individuals. This viral pathogen infects children more especially those under the age of 2 and may lead to death. It causes 3 million hospitalizations and up to 60,000 deaths annually for under the age of 5. The most vulnerable are immunocompromised individuals and asthmatic children with suboptimal antiviral defenses. It is associated with bronchiolitis, pneumonia, and bronchopneumonia. Despite all the current interventions and clinical trials, the only available therapeutic strategies for this viral infection are palliative care. Therefore, it is imperative to understand the pathogenicity of RSV and the corresponding host immune response to depict a sort of a targeted intervention. With the increasingly cutting-edge methods in harnessing the pathogenicity of this viral infection, high throughput systems including omics technological advances are at the spotlight. For instance, the associated genes with RSV complications for the host, the set of microbiome identified as operational taxonomic unit, the upregulated or downregulated metabolites, the protein subtypes, and the small molecules can help explain the viral microenvironment. Moreover, these big data will lead to RSV patients' stratification through individualized patient profiles that will bring in targeted prevention and treatment algorithms tailored to individualized patients' profiles. Through this, the virus and host interactions based on the pathogenicity of infection will provide a strong ground for depicting the prevention, prediction, and personalized medicine (3PM) for RSV. The 3PM approach brought cutting edge functional medicine to the healthcare givers, thus conferring targeted prevention and precision medicine while observing personalized treatment as well as preventive regularities. The viral replication mechanisms against the host defense mechanisms are crucial for the development of safe and effective therapy. Integrative personal omics profiles, whose analysis is based on the combined proteomics, transcriptomics, genomics, proteoformics, metabolomics, and autoantibody profiles, are very robust for predicting the risk of RSV infection. The targeted prevention will emerge from the patient stratification when the diagnosis is accurately predicted. In addition, the personalized medical services will give an effective prognostic assessment for RSV complications.
Collapse
Affiliation(s)
- Ousman Bajinka
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
2
|
Hägglund S, Laloy E, Alvarez I, Guo Y, Hallbrink Ågren G, Yazdan Panah H, Widgren A, Bergquist J, Hillström A, Baillif V, Saias L, Dubourdeau M, Timsit E, Valarcher JF. Effects of early treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) on the bronchoalveolar lavage proteome and oxylipids during bovine respiratory syncytial virus (BRSV) infection. PLoS One 2024; 19:e0309609. [PMID: 39546485 PMCID: PMC11567528 DOI: 10.1371/journal.pone.0309609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 11/17/2024] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAID) are not recommended for use against pneumonia in humans, but are commonly utilised against bovine respiratory disease. This study aimed to determine if the use of NSAIDs in the early phase of bovine respiratory syncytial virus (BRSV)-infection limits pulmonary inflammation. Four to nine-week old calves were infected with BRSV by aerosol and were treated with either meloxicam intravenously on day (D)4 (n = 5, MEL), acetylsalicylat-DL-lysin intravenously on D4 and D5 (n = 5, ASA), or were left untreated as controls (n = 5, CTR). Clinical signs were monitored daily until necropsy on D7, BRSV-RNA was detected in nasal swabs and bronchoalveolar lavage (BAL) by RT-qPCR, inflammatory cells and proteins were identified in BAL by cytology and label-free quantitative mass spectrometry-based proteomics, respectively, and oxylipids were quantified in BAL and plasma by liquid chromatography tandem mass spectrometry with triple quadrupole mass detectors. The calves developed mild to moderate signs of respiratory disease and, with the exception of one MEL-treated and one ASA-treated calf, limited lung lesions. None of the treatments had a significant effect on virus replication, clinical signs or lung lesion extent. Relative to controls, both treatments initially induced a downregulation of proteins in BAL. Immunoglobulin (Ig)-related proteins, such as the Ig kappa and lambda locus and the joining chain of IgA and IgM, were downregulated in MEL-treated calves compared to controls. In addition, meloxicam induced an increased neutrophil influx in BAL in response to BRSV, possibly related to a reduction in plasma prostaglandin, and to a downregulation of The Liver X Receptor/ Retinoid X Receptor (LXR/RXR), the Farnesoid X Receptor (FXR)/RXR and the 24-Dehydrocholesterol Reductase (DHC24) signalling pathways in the lung. The risk of NSAIDs to increase neutrophil activity during stimulation with BRSV or other toll-like receptor 4 agonists needs to be investigated further. Since augmented neutrophil responses can be detrimental, the results of the present study do not support the use of NSAIDs to prevent the clinical expression of BRSV-infection.
Collapse
Affiliation(s)
- Sara Hägglund
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Eve Laloy
- Laboratoire VETODIAG, Saint-Pierre-en-Auge, France
| | - Ignacio Alvarez
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Yongzhi Guo
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Gabriella Hallbrink Ågren
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Haleh Yazdan Panah
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Anna Widgren
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Anna Hillström
- Clinical Pathology Laboratory, University Animal Hospital, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | | | | | - Jean François Valarcher
- HPIG, Ruminant Medicine Unit, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
3
|
da Silva Barcelos L, Ford AK, Frühauf MI, Botton NY, Fischer G, Maggioli MF. Interactions Between Bovine Respiratory Syncytial Virus and Cattle: Aspects of Pathogenesis and Immunity. Viruses 2024; 16:1753. [PMID: 39599867 PMCID: PMC11598946 DOI: 10.3390/v16111753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Bovine respiratory syncytial virus (BRSV) is a major respiratory pathogen in cattle and is relevant to the livestock industry worldwide. BRSV is most severe in young calves and is often associated with stressful management events. The disease is responsible for economic losses due to lower productivity, morbidity, mortality, and prevention and treatment costs. As members of the same genus, bovine and human RSV share a high degree of homology and are similar in terms of their genomes, transmission, clinical signs, and epidemiology. This overlap presents an opportunity for One Health approaches and translational studies, with dual benefits; however, there is still a relative lack of studies focused on BRSV, and the continued search for improved prophylaxis highlights the need for a deeper understanding of its immunological features. BRSV employs different host-immunity-escaping mechanisms that interfere with effective long-term memory responses to current vaccines and natural infections. This review presents an updated description of BRSV's immunity processes, such as the PRRs and signaling pathways involved in BRSV infection, aspects of its pathogeny, and the evading mechanisms developed by the virus to thwart the immune response.
Collapse
Affiliation(s)
- Lariane da Silva Barcelos
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (L.d.S.B.)
- Laboratory of Virology and Immunology, Veterinary College, Universidade Federal de Pelotas, Capão do Leão, Rio Grande do Sul 96010, Brazil; (M.I.F.); (N.Y.B.); (G.F.)
| | - Alexandra K. Ford
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (L.d.S.B.)
| | - Matheus Iuri Frühauf
- Laboratory of Virology and Immunology, Veterinary College, Universidade Federal de Pelotas, Capão do Leão, Rio Grande do Sul 96010, Brazil; (M.I.F.); (N.Y.B.); (G.F.)
| | - Nadalin Yandra Botton
- Laboratory of Virology and Immunology, Veterinary College, Universidade Federal de Pelotas, Capão do Leão, Rio Grande do Sul 96010, Brazil; (M.I.F.); (N.Y.B.); (G.F.)
| | - Geferson Fischer
- Laboratory of Virology and Immunology, Veterinary College, Universidade Federal de Pelotas, Capão do Leão, Rio Grande do Sul 96010, Brazil; (M.I.F.); (N.Y.B.); (G.F.)
| | - Mayara Fernanda Maggioli
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (L.d.S.B.)
| |
Collapse
|
4
|
Alvarez I, Ducatez M, Guo Y, Lion A, Widgren A, Dubourdeau M, Baillif V, Saias L, Zohari S, Bergquist J, Meyer G, Valarcher JF, Hägglund S. Proteomic and Lipidomic Profiling of Calves Experimentally Co-Infected with Influenza D Virus and Mycoplasma bovis: Insights into the Host-Pathogen Interactions. Viruses 2024; 16:361. [PMID: 38543727 PMCID: PMC10975297 DOI: 10.3390/v16030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 05/23/2024] Open
Abstract
The role of Influenza D virus (IDV) in bovine respiratory disease remains unclear. An in vivo experiment resulted in increased clinical signs, lesions, and pathogen replication in calves co-infected with IDV and Mycoplasma bovis (M. bovis), compared to single-infected calves. The present study aimed to elucidate the host-pathogen interactions and profile the kinetics of lipid mediators in the airways of these calves. Bronchoalveolar lavage (BAL) samples collected at 2 days post-infection (dpi) were used for proteomic analyses by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Additionally, lipidomic analyses were performed by LC-MS/MS on BAL samples collected at 2, 7 and 14 dpi. Whereas M. bovis induced the expression of proteins involved in fibrin formation, IDV co-infection counteracted this coagulation mechanism and downregulated other acute-phase response proteins, such as complement component 4 (C4) and plasminogen (PLG). The reduced inflammatory response against M. bovis likely resulted in increased M. bovis replication and delayed M. bovis clearance, which led to a significantly increased abundance of oxylipids in co-infected calves. The identified induced oxylipids mainly derived from arachidonic acid; were likely oxidized by COX-1, COX-2, and LOX-5; and peaked at 7 dpi. This paper presents the first characterization of BAL proteome and lipid mediator kinetics in response to IDV and M. bovis infection in cattle and raises hypotheses regarding how IDV acts as a co-pathogen in bovine respiratory disease.
Collapse
Affiliation(s)
- Ignacio Alvarez
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Mariette Ducatez
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Yongzhi Guo
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Adrien Lion
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Anna Widgren
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden; (A.W.); (J.B.)
| | | | | | - Laure Saias
- Ambiotis SAS, 3 Rue des Satellites, 31400 Toulouse, France
| | - Siamak Zohari
- Department of Microbiology, Swedish Veterinary Agency, Ullsvägen 2B, 75189 Uppsala, Sweden;
| | - Jonas Bergquist
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden; (A.W.); (J.B.)
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Ulls väg 26, 75007 Uppsala, Sweden
| | - Gilles Meyer
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Jean-Francois Valarcher
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Sara Hägglund
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| |
Collapse
|
5
|
Díaz FE, McGill JL. Modeling Human Respiratory Syncytial Virus (RSV) Infection: Recent Contributions and Future Directions Using the Calf Model of Bovine RSV Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1180-1186. [PMID: 37782855 PMCID: PMC10558079 DOI: 10.4049/jimmunol.2300260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/24/2023] [Indexed: 10/04/2023]
Abstract
The human orthopneumovirus (human respiratory syncytial virus [RSV]) is a leading cause of respiratory disease in children worldwide and a significant cause of infant mortality in low- and middle-income countries. The natural immune response to the virus has a preponderant role in disease progression, with a rapid neutrophil infiltration and dysbalanced T cell response in the lungs associated with severe disease in infants. The development of preventive interventions against human RSV has been difficult partly due to the need to use animal models that only partially recapitulate the immune response as well as the disease progression seen in human infants. In this brief review, we discuss the contributions of the calf model of RSV infection to understanding immunity to RSV and in developing vaccine and drug candidates, focusing on recent research areas. We propose that the bovine model of RSV infection is a valuable alternative for assessing the translational potential of interventions aimed at the human population.
Collapse
Affiliation(s)
- Fabián E. Díaz
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
6
|
Zhang Y, Wang Q, Mackay CR, Ng LG, Kwok I. Neutrophil subsets and their differential roles in viral respiratory diseases. J Leukoc Biol 2022; 111:1159-1173. [PMID: 35040189 PMCID: PMC9015493 DOI: 10.1002/jlb.1mr1221-345r] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play significant roles in immune homeostasis and as neutralizers of microbial infections. Recent evidence further suggests heterogeneity of neutrophil developmental and activation states that exert specialized effector functions during inflammatory disease conditions. Neutrophils can play multiple roles during viral infections, secreting inflammatory mediators and cytokines that contribute significantly to host defense and pathogenicity. However, their roles in viral immunity are not well understood. In this review, we present an overview of neutrophil heterogeneity and its impact on the course and severity of viral respiratory infectious diseases. We focus on the evidence demonstrating the crucial roles neutrophils play in the immune response toward respiratory infections, using influenza as a model. We further extend the understanding of neutrophil function with the studies pertaining to COVID-19 disease and its neutrophil-associated pathologies. Finally, we discuss the relevance of these results for future therapeutic options through targeting and regulating neutrophil-specific responses.
Collapse
Affiliation(s)
- Yuning Zhang
- Department of ResearchNational Skin CentreSingaporeSingapore
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Charles R Mackay
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
- Department of Microbiology, Infection and Immunity ProgramBiomedicine Discovery Institute, Monash UniversityMelbourneAustralia
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
- State Key Laboratory of Experimental HematologyInstitute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Microbiology and ImmunologyImmunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of SingaporeSingaporeSingapore
- National Cancer Centre SingaporeSingaporeSingapore
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
| |
Collapse
|
7
|
Multipopulational transcriptome analysis of post-weaned beef cattle at arrival further validates candidate biomarkers for predicting clinical bovine respiratory disease. Sci Rep 2021; 11:23877. [PMID: 34903778 PMCID: PMC8669006 DOI: 10.1038/s41598-021-03355-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Bovine respiratory disease (BRD) remains the leading infectious disease in post-weaned beef cattle. The objective of this investigation was to contrast the at-arrival blood transcriptomes from cattle derived from two distinct populations that developed BRD in the 28 days following arrival versus cattle that did not. Forty-eight blood samples from two populations were selected for mRNA sequencing based on even distribution of development (n = 24) or lack of (n = 24) clinical BRD within 28 days following arrival; cattle which developed BRD were further stratified into BRD severity cohorts based on frequency of antimicrobial treatment: treated once (treated_1) or treated twice or more and/or died (treated_2+). Sequenced reads (~ 50 M/sample, 150 bp paired-end) were aligned to the ARS-UCD1.2 bovine genome assembly. One hundred and thirty-two unique differentially expressed genes (DEGs) were identified between groups stratified by disease severity (healthy, n = 24; treated_1, n = 13; treated_2+, n = 11) with edgeR (FDR ≤ 0.05). Differentially expressed genes in treated_1 relative to both healthy and treated_2+ were predicted to increase neutrophil activation, cellular cornification/keratinization, and antimicrobial peptide production. Differentially expressed genes in treated_2+ relative to both healthy and treated_1 were predicted to increase alternative complement activation, decrease leukocyte activity, and increase nitric oxide production. Receiver operating characteristic (ROC) curves generated from expression data for six DEGs identified in our current and previous studies (MARCO, CFB, MCF2L, ALOX15, LOC100335828 (aka CD200R1), and SLC18A2) demonstrated good-to-excellent (AUC: 0.800–0.899; ≥ 0.900) predictability for classifying disease occurrence and severity. This investigation identifies candidate biomarkers and functional mechanisms in at arrival blood that predicted development and severity of BRD.
Collapse
|
8
|
McDonald PO, Schill C, Maina TW, Samuel B, Porter M, Yoon I, McGill JL. Feeding Saccharomyces cerevisiae fermentation products lessens the severity of a viral-bacterial coinfection in preweaned calves. J Anim Sci 2021; 99:skab300. [PMID: 34673945 PMCID: PMC8599294 DOI: 10.1093/jas/skab300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/18/2021] [Indexed: 12/04/2022] Open
Abstract
We have previously reported that supplementation with Saccharomyces cerevisiae fermentation products (SCFP) ameliorates clinical signs and lung pathology following experimental bovine respiratory syncytial virus (BRSV) infection in preweaned dairy calves. The objectives of this study were to determine the effect of SCFP supplementation on the metabolic and endocrine responses, and disease outcome of a viral-bacterial coinfection in preweaned calves. Twenty-seven, 1- to 2-d-old Holstein-Angus cross calves were enrolled in the study; one SCFP calf was removed from the trial during the pre-challenge phase due to complications from nephritis. Calves were assigned to two treatment groups: control or SCFP-treated, base milk replacer with 1 g/d SCFP (Smartcare, soluble formula) and calf starter top dressed with 5 g/d SCFP (NutriTek, insoluble formula). Calves were infected with BRSV on day 21, followed 6 d later by intratracheal inoculation with Pasteurella multocida (PM). Calves were euthanized on day 10 post-viral infection. Calves receiving SCFP had reduced thoracic ultrasonography scores on day 7 post-viral infection (P = 0.03) and a tendency toward reduced scores on day 10 post-viral infection (P = 0.09). Calves receiving SCFP also had less severe lung pathology scores at necropsy (P = 0.06). No differences between treatments were observed in lung viral loads (P = 0.48) or bacterial lung recovery (P = 0.34); however, there was a distinction in the lung location for PM recovery, with PM isolated more frequently from the cranial lobes in SCFP-treated calves, but more frequently from the caudal lobes of control calves. Calves treated with SCFP tended (P = 0.07) to have higher serum IL-6 concentrations following the coinfection. Calves treated with SCFP had lower concentrations of serum nonesterified fatty acids and beta-hydroxybutyric acid compared with controls following experimental challenge (P = 0.03 and P = 0.08, respectively), suggesting metabolic changes favoring growth and development. There were no differences between groups in gene expression of insulin receptor, insulin-like growth factor 1 (IGF-1), IGF-1 receptor (IGF-1R), growth hormone receptor, or haptoglobin in the liver. Results from this study suggest that supplementing with SCFP may moderate the impact of a respiratory viral-bacterial coinfection on preweaned calves through metabolic and immune modifications.
Collapse
Affiliation(s)
- Paiton O McDonald
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| | - Courtney Schill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| | - Teresia W Maina
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| | - Beulah Samuel
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| | - Madison Porter
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| | - Ilkyu Yoon
- Diamond V Mills Inc., Cedar Rapids, IA 52404, USA
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010, USA
| |
Collapse
|
9
|
Johansson C, Kirsebom FCM. Neutrophils in respiratory viral infections. Mucosal Immunol 2021; 14:815-827. [PMID: 33758367 PMCID: PMC7985581 DOI: 10.1038/s41385-021-00397-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 02/04/2023]
Abstract
Viral respiratory infections are a common cause of severe disease, especially in infants, people who are immunocompromised, and in the elderly. Neutrophils, an important innate immune cell, infiltrate the lungs rapidly after an inflammatory insult. The most well-characterized effector mechanisms by which neutrophils contribute to host defense are largely extracellular and the involvement of neutrophils in protection from numerous bacterial and fungal infections is well established. However, the role of neutrophils in responses to viruses, which replicate intracellularly, has been less studied. It remains unclear whether and, by which underlying immunological mechanisms, neutrophils contribute to viral control or confer protection against an intracellular pathogen. Furthermore, neutrophils need to be tightly regulated to avoid bystander damage to host tissues. This is especially relevant in the lung where damage to delicate alveolar structures can compromise gas exchange with life-threatening consequences. It is inherently less clear how neutrophils can contribute to host immunity to viruses without causing immunopathology and/or exacerbating disease severity. In this review, we summarize and discuss the current understanding of how neutrophils in the lung direct immune responses to viruses, control viral replication and spread, and cause pathology during respiratory viral infections.
Collapse
Affiliation(s)
- Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | |
Collapse
|
10
|
Xie L, Ma Y, Opsomer G, Pascottini OB, Guan Y, Dong Q. Neutrophil extracellular traps in cattle health and disease. Res Vet Sci 2021; 139:4-10. [PMID: 34217982 DOI: 10.1016/j.rvsc.2021.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022]
Abstract
Neutrophils largely contribute to the first line of defense against the invasion of pathogens. They kill pathogens basically by the following mechanisms: phagocytosis and proteolytic degradation, the release of enzymes with bactericidal activities, and the production of fibers to entrap pathogens, also known as neutrophil extracellular traps (NETs). NETs capture pathogens as a mechanism of immune protection and have been studied in-depth in various fields of human medicine. However, research about NETs in cattle is relatively scarce. The present article reviews the generation mechanisms, structural composition, signal pathways, advantages (and disadvantages) of NETs, and summarizes the latest findings of NETs in cattle health and disease.
Collapse
Affiliation(s)
- Lei Xie
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yixiong Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Geert Opsomer
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Osvaldo Bogado Pascottini
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Yandong Guan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Qiang Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
11
|
Mann M, Brasier AR. Evolution of proteomics technologies for understanding respiratory syncytial virus pathogenesis. Expert Rev Proteomics 2021; 18:379-394. [PMID: 34018899 PMCID: PMC8277732 DOI: 10.1080/14789450.2021.1931130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/14/2021] [Indexed: 10/21/2022]
Abstract
Introduction: Respiratory syncytial virus (RSV) is a major human pathogen associated with long term morbidity. RSV replication occurs primarily in the epithelium, producing a complex cellular response associated with acute inflammation and long-lived changes in pulmonary function and allergic disease. Proteomics approaches provide important insights into post-transcriptional regulatory processes including alterations in cellular complexes regulating the coordinated innate response and epigenome.Areas covered: Peer-reviewed proteomics studies of host responses to RSV infections and proteomics techniques were analyzed. Methodologies identified include 1)." bottom-up" discovery proteomics, 2). Organellar proteomics by LC-gel fractionation; 3). Dynamic changes in protein interaction networks by LC-MS; and 4). selective reaction monitoring MS. We introduce recent developments in single-cell proteomics, top-down mass spectrometry, and photo-cleavable surfactant chemistries that will have impact on understanding how RSV induces extracellular matrix (ECM) composition and airway remodeling.Expert opinion: RSV replication induces global changes in the cellular proteome, dynamic shifts in nuclear proteins, and remodeling of epigenetic regulatory complexes linked to the innate response. Pathways discovered by proteomics technologies have led to deeper mechanistic understanding of the roles of heat shock proteins, redox response, transcriptional elongation complex remodeling and ECM secretion remodeling in host responses to RSV infections and pathological sequelae.
Collapse
Affiliation(s)
- Morgan Mann
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, USA
| | - Allan R Brasier
- Department of Internal Medicine and Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
12
|
Díaz FE, Guerra-Maupome M, McDonald PO, Rivera-Pérez D, Kalergis AM, McGill JL. A Recombinant BCG Vaccine Is Safe and Immunogenic in Neonatal Calves and Reduces the Clinical Disease Caused by the Respiratory Syncytial Virus. Front Immunol 2021; 12:664212. [PMID: 33981309 PMCID: PMC8108697 DOI: 10.3389/fimmu.2021.664212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) constitutes a major health burden, causing millions of hospitalizations in children under five years old worldwide due to acute lower respiratory tract infections. Despite decades of research, licensed vaccines to prevent hRSV are not available. Development of vaccines against hRSV targeting young infants requires ruling out potential vaccine-enhanced disease presentations. To achieve this goal, vaccine testing in proper animal models is essential. A recombinant BCG vaccine that expresses the Nucleoprotein of hRSV (rBCG-N-hRSV) protects mice against hRSV infection, eliciting humoral and cellular immune protection. Further, this vaccine was shown to be safe and immunogenic in human adult volunteers. Here, we evaluated the safety, immunogenicity, and protective efficacy of the rBCG-N-hRSV vaccine in a neonatal bovine RSV calf infection model. Newborn, colostrum-replete Holstein calves were either vaccinated with rBCG-N-hRSV, WT-BCG, or left unvaccinated, and then inoculated via aerosol challenge with bRSV strain 375. Vaccination with rBCG-N-hRSV was safe and well-tolerated, with no systemic adverse effects. There was no evidence of vaccine-enhanced disease following bRSV challenge of rBCG-N-hRSV vaccinated animals, suggesting that the vaccine is safe for use in neonates. Vaccination increased virus-specific IgA and virus-neutralization activity in nasal fluid and increased the proliferation of virus- and BCG-specific CD4+ and CD8+ T cells in PBMCs and lymph nodes at 7dpi. Furthermore, rBCG-N-hRSV vaccinated calves developed reduced clinical disease as compared to unvaccinated control calves, although neither pathology nor viral burden were significantly reduced in the lungs. These results suggest that the rBCG-N-hRSV vaccine is safe in neonatal calves and induces protective humoral and cellular immunity against this respiratory virus. These data from a newborn animal model provide further support to the notion that this vaccine approach could be considered as a candidate for infant immunization against RSV.
Collapse
Affiliation(s)
- Fabián E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Paiton O McDonald
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Daniela Rivera-Pérez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
13
|
Wallace LE, Liu M, van Kuppeveld FJM, de Vries E, de Haan CAM. Respiratory mucus as a virus-host range determinant. Trends Microbiol 2021; 29:983-992. [PMID: 33875348 PMCID: PMC8503944 DOI: 10.1016/j.tim.2021.03.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Efficient penetration of the mucus layer is needed for respiratory viruses to avoid mucociliary clearance prior to infection. Many respiratory viruses bind to glycans on the heavily glycosylated mucins that give mucus its gel-like characteristics. Influenza viruses, some paramyxoviruses, and coronaviruses avoid becoming trapped in the mucus by releasing themselves by means of their envelope-embedded enzymes that destroy glycan receptors. For efficient infection, receptor binding and destruction need to be in balance with the host receptor repertoire. Establishment in a novel host species requires resetting of the balance to adapt to the different glycan repertoire encountered. Growing understanding of species-specific mucosal glycosylation patterns and the dynamic interaction with respiratory viruses identifies the mucus layer as a major host-range determinant and barrier for zoonotic transfer.
Collapse
Affiliation(s)
- Louisa E Wallace
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Mengying Liu
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Frank J M van Kuppeveld
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Erik de Vries
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands.
| | - Cornelis A M de Haan
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Single-Shot Vaccines against Bovine Respiratory Syncytial Virus (BRSV): Comparative Evaluation of Long-Term Protection after Immunization in the Presence of BRSV-Specific Maternal Antibodies. Vaccines (Basel) 2021; 9:vaccines9030236. [PMID: 33803302 PMCID: PMC8001206 DOI: 10.3390/vaccines9030236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 11/28/2022] Open
Abstract
The induction of long-lasting clinical and virological protection is needed for a successful vaccination program against the bovine respiratory syncytial virus (BRSV). In this study, calves with BRSV-specific maternally derived antibodies were vaccinated once, either with (i) a BRSV pre-fusion protein (PreF) and MontanideTM ISA61 VG (ISA61, n = 6), (ii) BRSV lacking the SH gene (ΔSHrBRSV, n = 6), (iii) a commercial vaccine (CV, n = 6), or were injected with ISA61 alone (n = 6). All calves were challenged with BRSV 92 days later and were euthanized 13 days post-infection. Based on clinical, pathological, and proteomic data, all vaccines appeared safe. Compared to the controls, PreF induced the most significant clinical and virological protection post-challenge, followed by ΔSHrBRSV and CV, whereas the protection of PreF-vaccinated calves was correlated with BRSV-specific serum immunoglobulin (Ig)G antibody responses 84 days post-vaccination, and the IgG antibody titers of ΔSHrBRSV- and CV-vaccinated calves did not differ from the controls on this day. Nevertheless, strong anamnestic BRSV- and PreF-specific IgG responses occurred in calves vaccinated with either of the vaccines, following a BRSV challenge. In conclusion, PreF and ΔSHrBRSV are two efficient one-shot candidate vaccines. By inducing a protection for at least three months, they could potentially improve the control of BRSV in calves.
Collapse
|
15
|
Bassel LL, Kaufman EI, Alsop SNA, Buchan J, Hewson J, McCandless EE, Tiwari R, Sharif S, Vulikh K, Caswell JL. Effect of aerosolized bacterial lysate on development of naturally occurring respiratory disease in beef calves. J Vet Intern Med 2021; 35:655-665. [PMID: 33442910 PMCID: PMC7848379 DOI: 10.1111/jvim.16032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/14/2023] Open
Abstract
Background Bovine respiratory disease (BRD) is a major problem affecting beef cattle after arrival to feedlots. Alternatives to antibiotics are needed for prevention. Hypothesis Stimulation of pulmonary innate immune responses at the time of arrival to a feedlot reduces the occurrence and severity of BRD. Animals Sixty beef steers at high risk of BRD. Methods Randomized, double‐blinded, placebo‐controlled study. Calves received saline or a lysate of Staphylococcus aureus and Escherichia coli by aerosol, at 16 hours after feedlot arrival. Calves were monitored for 28 days for disease outcomes and levels of Mycoplasma bovis and Mannheimia haemolytica in nasal swabs. Results Death from M bovis pneumonia was significantly greater in lysate‐treated animals (6/29, 24%) compared to controls (1/29, 3%; odds ratio = 10.2; 95% confidence interval [CI] = 1.1‐96.0; P = .04). By 28 days after arrival, 29/29 lysate‐treated calves had ultrasonographic pulmonary consolidation compared to 24/29 control calves (P = .05). Lysate‐treated calves had lower weight gain compared to control calves (−8.8 kg, 95% CI = −17.1 to −0.5; P = .04), and higher body temperatures on days 4, 7, and 21 (0.19°C; 95% CI = 0.01‐0.37; P = .04). Nasal M bovis numbers increased over time and were higher in lysate‐treated calves (0.76 log CFU, 95% CI = 0.3‐1.2; P = .001). Conclusions and Clinical Importance Aerosol administration of a bacterial lysate exacerbated BRD in healthy high‐risk beef calves, suggesting that respiratory tract inflammation adversely affects how calves respond to subsequent natural infection with M bovis and other respiratory pathogens.
Collapse
Affiliation(s)
- Laura L Bassel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Emily I Kaufman
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Sarah-Nicole A Alsop
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jordan Buchan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Joanne Hewson
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Erin E McCandless
- Global Therapeutics Research, Veterinary Medicine Research and Development, Zoetis Inc., Kalamazoo, Michigan, USA
| | - Raksha Tiwari
- Global Therapeutics Research, Veterinary Medicine Research and Development, Zoetis Inc., Kalamazoo, Michigan, USA
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ksenia Vulikh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jeff L Caswell
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
16
|
Mahmoud AHA, Slate JR, Hong S, Yoon I, McGill JL. Supplementing a Saccharomyces cerevisiae fermentation product modulates innate immune function and ameliorates bovine respiratory syncytial virus infection in neonatal calves. J Anim Sci 2020; 98:5891219. [PMID: 32780814 PMCID: PMC7457959 DOI: 10.1093/jas/skaa252] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/03/2020] [Indexed: 12/01/2022] Open
Abstract
The objectives of this study were to determine the effects of oral supplementation with Saccharomyces cerevisiae fermentation products (SCFP; SmartCare and NutriTek; Diamond V, Cedar Rapids, IA) on immune function and bovine respiratory syncytial virus (BRSV) infection in preweaned dairy calves. Twenty-four Holstein × Angus, 1- to 2-d-old calves (38.46 ± 0.91 kg initial body weight [BW]) were assigned two treatment groups: control or SCFP treated, milk replacer with 1 g/d SCFP (SmartCare) and calf starter top-dressed with 5 g/d SCFP (NutriTek). The study consisted of one 31-d period. On days 19 to 21 of the supplementation period, calves were challenged via aerosol inoculation with BRSV strain 375. Calves were monitored twice daily for clinical signs, including rectal temperature, cough, nasal and ocular discharge, respiration effort, and lung auscultation. Calves were euthanized on day 10 postinfection (days 29 to 31 of the supplementation period) to evaluate gross lung pathology and pathogen load. Supplementation with SCFP did not affect BW (P = 0.762) or average daily gain (P = 0.750), percentages of circulating white blood cells (P < 0.05), phagocytic (P = 0.427 for neutrophils and P = 0.460 for monocytes) or respiratory burst (P = 0.119 for neutrophils and P = 0.414 for monocytes) activity by circulating leukocytes either before or following BRSV infection, or serum cortisol concentrations (P = 0.321) after BRSV infection. Calves receiving SCFP had reduced clinical disease scores compared with control calves (P = 0.030), reduced airway neutrophil recruitment (P < 0.002), reduced lung pathology (P = 0.031), and a reduced incidence of secondary bacterial infection. Calves receiving SCFP shed reduced virus compared with control calves (P = 0.049) and tended toward lower viral loads in the lungs (P = 0.051). Immune cells from the peripheral blood of SCFP-treated calves produced increased (P < 0.05) quantities of interleukin (IL)-6 and tumor necrosis factor-alpha in response to toll-like receptor stimulation, while cells from the bronchoalveolar lavage (BAL) of SCFP-treated calves secreted less (P < 0.05) proinflammatory cytokines in response to the same stimuli. Treatment with SCFP had no effect on virus-specific T cell responses in the blood but resulted in reduced (P = 0.045) virus-specific IL-17 secretion by T cells in the BAL. Supplementing with SCFP modulates both systemic and mucosal immune responses and may improve the outcome of an acute respiratory viral infection in preweaned dairy calves.
Collapse
Affiliation(s)
- Asmaa H A Mahmoud
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA.,Agricultural Research Center, Animal Health Research Institute, Giza, Egypt
| | - Jamison R Slate
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA
| | - Suyeon Hong
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA
| | | | - Jodi L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA
| |
Collapse
|
17
|
Byrne KA, Loving CL, McGill JL. Innate Immunomodulation in Food Animals: Evidence for Trained Immunity? Front Immunol 2020; 11:1099. [PMID: 32582185 PMCID: PMC7291600 DOI: 10.3389/fimmu.2020.01099] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/06/2020] [Indexed: 01/07/2023] Open
Abstract
Antimicrobial resistance (AMR) is a significant problem in health care, animal health, and food safety. To limit AMR, there is a need for alternatives to antibiotics to enhance disease resistance and support judicious antibiotic usage in animals and humans. Immunomodulation is a promising strategy to enhance disease resistance without antibiotics in food animals. One rapidly evolving field of immunomodulation is innate memory in which innate immune cells undergo epigenetic changes of chromatin remodeling and metabolic reprogramming upon a priming event that results in either enhanced or suppressed responsiveness to secondary stimuli (training or tolerance, respectively). Exposure to live agents such as bacille Calmette-Guerin (BCG) or microbe-derived products such as LPS or yeast cell wall ß-glucans can reprogram or "train" the innate immune system. Over the last decade, significant advancements increased our understanding of innate training in humans and rodent models, and strategies are being developed to specifically target or regulate innate memory. In veterinary species, the concept of enhancing the innate immune system is not new; however, there are few available studies which have purposefully investigated innate training as it has been defined in human literature. The development of targeted approaches to engage innate training in food animals, with the practical goal of enhancing the capacity to limit disease without the use of antibiotics, is an area which deserves attention. In this review, we provide an overview of innate immunomodulation and memory, and the mechanisms which regulate this long-term functional reprogramming in other animals (e.g., humans, rodents). We focus on studies describing innate training, or similar phenomenon (often referred to as heterologous or non-specific protection), in cattle, sheep, goats, swine, poultry, and fish species; and discuss the potential benefits and shortcomings of engaging innate training for enhancing disease resistance.
Collapse
Affiliation(s)
- Kristen A. Byrne
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Services, USDA, Ames, IA, United States
| | - Crystal L. Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Services, USDA, Ames, IA, United States
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Riffault S, Hägglund S, Guzman E, Näslund K, Jouneau L, Dubuquoy C, Pietralunga V, Laubreton D, Boulesteix O, Gauthier D, Remot A, Boukaridi A, Falk A, Shevchenko G, Lind SB, Vargmar K, Zhang B, Kwong PD, Rodriguez MJ, Duran MG, Schwartz-Cornil I, Eléouët JF, Taylor G, Valarcher JF. A Single Shot Pre-fusion-Stabilized Bovine RSV F Vaccine is Safe and Effective in Newborn Calves with Maternally Derived Antibodies. Vaccines (Basel) 2020; 8:vaccines8020231. [PMID: 32443437 PMCID: PMC7349975 DOI: 10.3390/vaccines8020231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 01/21/2023] Open
Abstract
Achieving safe and protective vaccination against respiratory syncytial virus (RSV) in infants and in calves has proven a challenging task. The design of recombinant antigens with a conformation close to their native form in virus particles is a major breakthrough. We compared two subunit vaccines, the bovine RSV (BRSV) pre-fusion F (preF) alone or with nanorings formed by the RSV nucleoprotein (preF+N). PreF and N proteins are potent antigenic targets for neutralizing antibodies and T cell responses, respectively. To tackle the challenges of neonatal immunization, three groups of six one-month-old calves with maternally derived serum antibodies (MDA) to BRSV received a single intramuscular injection of PreF, preF+N with MontanideTM ISA61 VG (ISA61) as adjuvant or only ISA61 (control). One month later, all calves were challenged with BRSV and monitored for virus replication in the upper respiratory tract and for clinical signs of disease over one week, and then post-mortem examinations of their lungs were performed. Both preF and preF+N vaccines afforded safe, clinical, and virological protection against BRSV, with little difference between the two subunit vaccines. Analysis of immune parameters pointed to neutralizing antibodies and antibodies to preF as being significant correlates of protection. Thus, a single shot vaccination with preF appears sufficient to reduce the burden of BRSV disease in calves with MDA.
Collapse
Affiliation(s)
- Sabine Riffault
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
- Correspondence: ; Tel.: +33-(0)-134-652-620
| | - Sara Hägglund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Efrain Guzman
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Katarina Näslund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Luc Jouneau
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Catherine Dubuquoy
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Vincent Pietralunga
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Daphné Laubreton
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | | | | | - Aude Remot
- INRAE, University of Tours, ISP, 37380 Nouzilly, France;
| | - Abdelhak Boukaridi
- University Paris Saclay, INRAE, AgroParisTech, GABI, 78350 Jouy-en-Josas, France;
| | - Alexander Falk
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Ganna Shevchenko
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Sara Bergström Lind
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Karin Vargmar
- Department of Biomedicine and veterinary public Health, Swedish University of Agricultural Sciences, Box 7054, SE-756 51, 875007 Uppsala, Sweden;
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - María Jose Rodriguez
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Marga Garcia Duran
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Isabelle Schwartz-Cornil
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Jean-François Eléouët
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Jean François Valarcher
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| |
Collapse
|
19
|
McGill JL, Sacco RE. The Immunology of Bovine Respiratory Disease: Recent Advancements. Vet Clin North Am Food Anim Pract 2020; 36:333-348. [PMID: 32327252 PMCID: PMC7170797 DOI: 10.1016/j.cvfa.2020.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, 1907 ISU C-Drive, VMRI Building 5, Ames, IA 50010, USA.
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, Agricultural Research Services, USDA, PO Box 70, 1920 Dayton Avenue, Ames, IA 50010, USA
| |
Collapse
|
20
|
Aljabr W, Armstrong S, Rickett NY, Pollakis G, Touzelet O, Cloutman-Green E, Matthews DA, Hiscox JA. High Resolution Analysis of Respiratory Syncytial Virus Infection In Vivo. Viruses 2019; 11:v11100926. [PMID: 31658630 PMCID: PMC6832471 DOI: 10.3390/v11100926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/27/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is a major cause of pediatric infection and also causes disease in the elderly and those with underlying respiratory problems. There is no vaccine for HRSV and anti-viral therapeutics are not broadly applicable. To investigate the effect of HRSV biology in children, nasopharyngeal aspirates were taken from children with different viral loads and a combined high throughput RNAseq and label free quantitative proteomics approach was used to characterize the nucleic acid and proteins in these samples. HRSV proteins were identified in the nasopharyngeal aspirates from infected children, and their abundance correlated with viral load (Ct value), confirming HRSV infection. Analysis of the HRSV genome indicated that the children were infected with sub-group A virus and that minor variants in nucleotide frequency occurred in discrete clusters along the HRSV genome, and within a patient clustered distinctly within the glycoprotein gene. Data from the samples were binned into four groups; no-HRSV infection (control), high viral load (Ct < 20), medium viral load (Ct = 20-25), and low viral load (Ct > 25). Cellular proteins associated with the anti-viral response (e.g., ISG15) were identified in the nasopharyngeal aspirates and their abundance was correlated with viral load. These combined approaches have not been used before to study HRSV biology in vivo and can be readily applied to the study the variation of virus host interactions.
Collapse
Affiliation(s)
- Waleed Aljabr
- King Fahad Medical City, Research Center, 59046 Riyadh 11525, Saudi Arabia.
| | - Stuart Armstrong
- Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK.
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool L3 5RF, UK.
| | - Natasha Y Rickett
- Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK.
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool L3 5RF, UK.
| | - Georgios Pollakis
- Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK.
| | - Olivier Touzelet
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, UK.
| | | | - David A Matthews
- School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | - Julian A Hiscox
- Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK.
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool L3 5RF, UK.
| |
Collapse
|
21
|
Hofstetter AR, Sacco RE. Oxidative stress pathway gene transcription after bovine respiratory syncytial virus infection in vitro and ex vivo. Vet Immunol Immunopathol 2019; 219:109956. [PMID: 31706084 DOI: 10.1016/j.vetimm.2019.109956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 11/26/2022]
Abstract
Studies in mouse and lamb models indicate important roles of reactive oxygen species (ROS) in the pathology and immune response to respiratory syncytial virus (RSV). The role of ROS in bovine RSV (BRSV) infection of calves remains unclear. BRSV naturally infects calves, leading to similar disease course, micro- and macro-lesions, and symptomology as is observed in RSV infection of human neonates. Furthermore, humans, lambs, and calves, but not mice, have an active lung oxidative system involving lactoperoxidase (LPO) and the dual oxidases (DUOX) 1 and 2. To gain insight into the role of ROS in the BRSV-infected lung, we examined gene expression in infected bovine cells using qPCR. A panel of 19 primers was used to assay ex vivo and in vitro BRSV-infected cells. The panel targeted genes involved in both production and regulation of ROS. BRSV infection significantly increased transcription of five genes in bovine respiratory tract cells in vitro and ex vivo. PTGS2 expression more than doubled in both sample types. Four transcripts varied significantly in lung lesions, but not non-lesion samples, compared with uninfected lung. This is the first report of the transcriptional profile of ROS-related genes in the airway after BRSV infection in the natural host.
Collapse
Affiliation(s)
- Amelia R Hofstetter
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, United States Department of Agriculture, 1920 Dayton Avenue, Ames, IA, 50010, United States of America.
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, United States Department of Agriculture, 1920 Dayton Avenue, Ames, IA, 50010, United States of America.
| |
Collapse
|
22
|
Chézeau L, Kohlstaedt LA, Le Faou A, Cosnier F, Rihn B, Gaté L. Proteomic analysis of bronchoalveolar lavage fluid in rat exposed to TiO 2 nanostructured aerosol by inhalation. J Proteomics 2019; 207:103451. [PMID: 31323425 DOI: 10.1016/j.jprot.2019.103451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/23/2019] [Accepted: 07/15/2019] [Indexed: 12/30/2022]
Abstract
The pulmonary toxicological properties of inhaled titanium dioxide were studied using bronchoalveolar lavage fluid (BALF) cytology and proteomics analyses. Fischer 344 rats were exposed to 10 mg/m3 of TiO2 nanostructured aerosol by nose-only inhalation for 6 h/day, 5 days/week for 4 weeks. Lung samples were collected up to 180 post-exposure days. As previously described, cytological analyses of BALF showed a strong inflammatory response up to 3 post-exposure days, which persisted however, at a lower intensity up to 180 days. In addition, using Multidimensional Protein Identification Technology (MudPIT), we identified a total of 107, 50 and 45 proteins (UniprotKB identifiers) differentially expressed in exposed rats immediately, 3 and 180 days after the end of exposure respectively. Increased levels of inflammatory proteins, members of proteasome, various histones, proteins involved in cytoskeleton organization, were noticed up to 3 days (short-term response). Some of these proteins were linked with Neutrophil Extracellular Trap formation (NETosis). Long-term response was also characterized by a persistent altered expression of proteins up to 180 days. Altogether, these results suggest that exposure to low toxicity low solubility nanomaterials such as TiO2 may induce long-term changes in the pulmonary protein expression pattern of which the physio-pathological consequences are unknown. SIGNIFICANCE: This paper describes in rats, at the pulmonary level, the effects of inhaled nanostructured aerosol of TiO2 on the secreted proteins found in the broncho-alveolar space by comparing the proteomic profile in broncho-alveolar lavage fluid supernatants of control and exposed animals. This work brings new insights about the early events occurring following the end of exposure and suggests the formation of Neutrophil Extracellular Traps (NETosis) that could be interpret as a potential early mechanism of defense against TiO2 nanoparticles. This work also describes the long term effects (180 post-exposure days) of such an exposure and the change in secreted protein expression in the absence of significant histopathological modifications.
Collapse
Affiliation(s)
- Laëtitia Chézeau
- Institut National de Recherche et de Sécurité, Rue du Morvan, CS 60027, 54519 Vandœuvre, Cedex, France; EA 3452 CITHEFOR, Université de Lorraine, BP 80403, 54001 Nancy Cedex, France
| | - Lori A Kohlstaedt
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, United States of America
| | - Alain Le Faou
- EA 3452 CITHEFOR, Université de Lorraine, BP 80403, 54001 Nancy Cedex, France
| | - Frédéric Cosnier
- Institut National de Recherche et de Sécurité, Rue du Morvan, CS 60027, 54519 Vandœuvre, Cedex, France
| | - Bertrand Rihn
- EA 3452 CITHEFOR, Université de Lorraine, BP 80403, 54001 Nancy Cedex, France; Institut Jean-Lamour, UMR 7198 CNRS, Université de Lorraine, 54011 Nancy Cedex, France
| | - Laurent Gaté
- Institut National de Recherche et de Sécurité, Rue du Morvan, CS 60027, 54519 Vandœuvre, Cedex, France.
| |
Collapse
|
23
|
Profiling of local disease-sparing responses to bovine respiratory syncytial virus in intranasally vaccinated and challenged calves. J Proteomics 2019; 204:103397. [PMID: 31146050 DOI: 10.1016/j.jprot.2019.103397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
Bovine and human respiratory syncytial viruses (BRSV, HRSV) are primary causes of pneumonia in calves and children respectively, with vaccination offering protection via antibody and cellular immune responses. However, with no vaccines currently licensed for human use, evaluation of local responses to BRSV vaccination may provide insights to aid the design of effective safe HRSV vaccines. Calves received intranasal single component BRSV vaccine or "3-Way" vaccine (BRSV, Bovine Herpes Virus-1 (BHV-1), Bovine Parainfluenza Virus Type-3 (BPIV-3)), and were BRSV-challenged 42 days post-vaccination. All vaccinates exhibited reduced pulmonary lesioning with elevated anti-BRSV serum IgG, and higher nasal anti-BRSV IgA in 3-Way vaccinates. Thirty-nine proteins associated with homeostatic and immune processes were altered in vaccinates, with enhanced 3-Way vaccinate group proteins associated with Th1/Th2 balance and immunoglobulin class switching. Proteins altered in the pharyngeal tonsil of animals euthanized early related to anti-inflammatory responses and lymphoid tissue remodeling. These findings indicate that multivalent vaccines distinctly modulate local immune responses, with clear correlation between the pharyngeal tonsil proteome profile and resulting immune protection and disease-sparing. This suggests that the efficacy of low-antigenic subunit vaccine components for problematic pathogens such as HRSV could be enhanced by use in combination with existing safe live vaccines. SIGNIFICANCE: This study demonstrates that vaccine valency can alter post-challenge proteome responses within the pharyngeal tonsil, a sentinel site of primary immune responses, with the magnitude of response dependent on antigen formulation. Observed differential responses can be attributed to antigenic material and viral nucleic acid from multivalent formulations providing additional T-cell epitopes and PAMPS. These findings indicate that incorporation of subunits proteins within multivalent formulations containing live virus has the potential to induce/skew a favorable immune response, utilising the natural adjuvanting effects of safe proven live vaccines.
Collapse
|
24
|
Zhong C, Li J, Mao L, Liu M, Zhu X, Li W, Sun M, Ji X, Xiao F, Yang L, Zhang W, Liao Z. Proteomics analysis reveals heat shock proteins involved in caprine parainfluenza virus type 3 infection. BMC Vet Res 2019; 15:151. [PMID: 31101113 PMCID: PMC6525452 DOI: 10.1186/s12917-019-1897-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/01/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Caprine parainfluenza virus type 3 (CPIV3) is major pathogen of goat herds causing serious respiratory tract disease and economic losses to the goat industry in China. We analyzed the differential proteomics of CPIV3-infected Madin-Darby bovine kidney (MDBK) cells using quantitative iTRAQ coupled LC-MS/MS. In addition, four DEPs were validated by qRT-PCR and western blot analysis. RESULTS Quantitative proteomics analysis revealed 163 differentially expressed proteins (DEPs) between CPIV3-infected and mock-infected groups (p-value < 0.05 and fold change > 1.2), among which 91 were down-regulated and 72 were up-regulated. Gene ontology (GO) analysis showed that these DEPs were involved in molecular functions, cellular components and biological processes. Biological functions in which the DEPs were involved in included diseases, genetic information processing, metabolism, environmental information processing, cellular processes, and organismal systems. STRING analysis revealed that four heat shock proteins (HSPs) included HSPA5, HSPA1B, HSP90B1 and HSPA6 may be associated with proliferation of CPIV3 in MDBK cells. qRT-PCR and western blot analysis showed that the selected HSPs were identical to the quantitative proteomics data. CONCLUSION To our knowledge, this is the first report of the proteomic changes in MDBK cells after CPIV3 infection.
Collapse
Affiliation(s)
- Chunyan Zhong
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.,College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China. .,School of Pharmacy, Linyi University, Linyi, 276000, China.
| | - Li Mao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.,Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Nanjing, 210014, China
| | - Maojun Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Xing Zhu
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Wenliang Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Min Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Xinqin Ji
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Fang Xiao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.,College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Leilei Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Wenwen Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Zheng Liao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.,College of Animal Science, Guizhou University, Guiyang, 550025, China
| |
Collapse
|
25
|
Altamirano-Lagos MJ, Díaz FE, Mansilla MA, Rivera-Pérez D, Soto D, McGill JL, Vasquez AE, Kalergis AM. Current Animal Models for Understanding the Pathology Caused by the Respiratory Syncytial Virus. Front Microbiol 2019; 10:873. [PMID: 31130923 PMCID: PMC6510261 DOI: 10.3389/fmicb.2019.00873] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the main etiologic agent of severe lower respiratory tract infections that affect young children throughout the world, associated with significant morbidity and mortality, becoming a serious public health problem globally. Up to date, no licensed vaccines are available to prevent severe hRSV-induced disease, and the generation of safe-effective vaccines has been a challenging task, requiring constant biomedical research aimed to overcome this ailment. Among the difficulties presented by the study of this pathogen, it arises the fact that there is no single animal model that resembles all aspects of the human pathology, which is due to the specificity that this pathogen has for the human host. Thus, for the study of hRSV, different animal models might be employed, depending on the goal of the study. Of all the existing models, the murine model has been the most frequent model of choice for biomedical studies worldwide and has been of great importance at contributing to the development and understanding of vaccines and therapies against hRSV. The most notable use of the murine model is that it is very useful as a first approach in the development of vaccines or therapies such as monoclonal antibodies, suggesting in this way the direction that research could have in other preclinical models that have higher maintenance costs and more complex requirements in its management. However, several additional different models for studying hRSV, such as other rodents, mustelids, ruminants, and non-human primates, have been explored, offering advantages over the murine model. In this review, we discuss the various applications of animal models to the study of hRSV-induced disease and the advantages and disadvantages of each model, highlighting the potential of each model to elucidate different features of the pathology caused by the hRSV infection.
Collapse
Affiliation(s)
- María José Altamirano-Lagos
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián E. Díaz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Andrés Mansilla
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Abel E. Vasquez
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
26
|
Meade KG, O'Farrelly C. β-Defensins: Farming the Microbiome for Homeostasis and Health. Front Immunol 2019; 9:3072. [PMID: 30761155 PMCID: PMC6362941 DOI: 10.3389/fimmu.2018.03072] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/11/2018] [Indexed: 12/18/2022] Open
Abstract
Diverse commensal populations are now regarded as key to physiological homeostasis and protection against disease. Although bacteria are the most abundant component of microbiomes, and the most intensively studied, the microbiome also consists of viral, fungal, archael, and protozoan communities, about which comparatively little is known. Host-defense peptides (HDPs), originally described as antimicrobial, now have renewed significance as curators of the pervasive microbial loads required to maintain homeostasis and manage microbiome diversity. Harnessing HDP biology to transition away from non-selective, antibiotic-mediated treatments for clearance of microbes is a new paradigm, particularly in veterinary medicine. One family of evolutionarily conserved HDPs, β-defensins which are produced in diverse combinations by epithelial and immune cell populations, are multifunctional cationic peptides which manage the cross-talk between host and microbes and maintain a healthy yet dynamic equilibrium across mucosal systems. They are therefore key gatekeepers to the oral, respiratory, reproductive and enteric tissues, preventing pathogen-associated inflammation and disease and maintaining physiological normality. Expansions in the number of genes encoding these natural antibiotics have been described in the genomes of some species, the functional significance of which has only recently being appreciated. β-defensin expression has been documented pre-birth and disruptions in their regulation may play a role in maladaptive neonatal immune programming, thereby contributing to subsequent disease susceptibility. Here we review recent evidence supporting a critical role for β-defensins as farmers of the pervasive and complex prokaryotic ecosystems that occupy all body surfaces and cavities. We also share some new perspectives on the role of β-defensins as sensors of homeostasis and the immune vanguard particularly at sites of immunological privilege where inflammation is attenuated.
Collapse
Affiliation(s)
- Kieran G. Meade
- Animal and Bioscience Research Centre, Teagasc, Grange, Ireland
| | - Cliona O'Farrelly
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Guerra-Maupome M, Palmer MV, McGill JL, Sacco RE. Utility of the Neonatal Calf Model for Testing Vaccines and Intervention Strategies for Use against Human RSV Infection. Vaccines (Basel) 2019; 7:vaccines7010007. [PMID: 30626099 PMCID: PMC6466205 DOI: 10.3390/vaccines7010007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of pediatric respiratory tract infections. It is estimated that two-thirds of infants are infected with RSV during the first year of life and it is one of the leading causes of death in this age group worldwide. Similarly, bovine RSV is a primary viral pathogen in cases of pneumonia in young calves and plays a significant role in bovine respiratory disease complex. Importantly, naturally occurring infection of calves with bovine RSV shares many features in common with human RSV infection. Herein, we update our current understanding of RSV infection in cattle, with particular focus on similarities between the calf and human infection, and the recent reports in which the neonatal calf has been employed for the development and testing of vaccines and therapeutics which may be applied to hRSV infection in humans.
Collapse
Affiliation(s)
- Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Mitchell V Palmer
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| |
Collapse
|