1
|
Joyce JD, Moore GA, Thompson CK, Bertke AS. Guinea Pigs Are Not a Suitable Model to Study Neurological Impacts of Ancestral SARS-CoV-2 Intranasal Infection. Viruses 2025; 17:706. [PMID: 40431717 PMCID: PMC12116045 DOI: 10.3390/v17050706] [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: 03/29/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Neurological symptoms involving the central nervous system (CNS) and peripheral nervous system (PNS) are common complications of acute COVID-19 as well as post-COVID conditions. Most research into these neurological sequalae focuses on the CNS, disregarding the PNS. Guinea pigs were previously shown to be useful models of disease during the SARS-CoV-1 epidemic. However, their suitability for studying SARS-CoV-2 has not been experimentally demonstrated. To assess the suitability of guinea pigs as models for SARS-CoV-2 infection and the impact of SARS-CoV-2 infection on the PNS, and to determine routes of CNS invasion through the PNS, we intranasally infected wild-type Dunkin-Hartley guinea pigs with ancestral SARS-CoV-2 USA-WA1/2020. We assessed PNS sensory neurons (trigeminal ganglia, dorsal root ganglia), autonomic neurons (superior cervical ganglia), brain regions (olfactory bulb, brainstem, cerebellum, cortex, hippocampus), lungs, and blood for viral RNA (RT-qPCR), protein (immunostaining), and infectious virus (plaque assay) at three- and six-days post infection. We show that guinea pigs, which have previously been used as a model of SARS-CoV-1 pulmonary disease, are not susceptible to intranasal infection with ancestral SARS-CoV-2, and are not useful models in assessing neurological impacts of infection with SARS-CoV-2 isolates from the early pandemic.
Collapse
Affiliation(s)
- Jonathan D. Joyce
- Translational Biology, Medicine and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA;
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Greyson A. Moore
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Christopher K. Thompson
- School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Andrea S. Bertke
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| |
Collapse
|
2
|
Harriott NC, Chimenti MS, Bonde G, Ryan AL. MixOmics Integration of Biological Datasets Identifies Highly Correlated Variables of COVID-19 Severity. Int J Mol Sci 2025; 26:4743. [PMID: 40429886 PMCID: PMC12111767 DOI: 10.3390/ijms26104743] [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: 12/19/2024] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Despite several years passing since the COVID-19 pandemic was declared, challenges remain in understanding the factors that can predict the severity of COVID-19 disease and complications of SARS-CoV-2 infection. While many large-scale multi-omic datasets have been published, integration of these datasets has the potential to substantially increase the biological insight gained, allowing a more complex comprehension of the disease pathogenesis. Such insight may improve our ability to predict disease progression, detect severe cases more rapidly and develop effective therapeutics. In this study, we have applied an innovative machine learning algorithm to delineate COVID severity based on the integration of paired samples of proteomic and transcriptomic data from a small cohort of patients testing positive for SARS-CoV-2 infection with differential disease severity. Targeted plasma proteomics and an onco-immune targeted transcriptomic panel were performed on sequential samples from a cohort of 23 severe, 21 moderate and 10 mild COVID-19 patients. We applied DIABLO, a new integrative method, to identify multi-omics biomarker panels that can discriminate between multiple phenotypic groups, such as the varied severity of disease in COVID-19 patients. As COVID-19 severity is known among our sample group, we can train models using this as the outcome variable and calculate features that are important predictors of severe disease. In this study, we detect highly correlated key variables of severe COVID-19 using transcriptomic discriminant analysis and multi-omics integration methods. This approach highlights the power of data integration from a small cohort of patients, offering a better biological understanding of the molecular mechanisms driving COVID-19 severity and an opportunity to improve the prediction of disease trajectories and targeted therapeutics.
Collapse
Affiliation(s)
- Noa C. Harriott
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA; (N.C.H.); (G.B.)
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael S. Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA;
| | - Gregory Bonde
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA; (N.C.H.); (G.B.)
| | - Amy L. Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA; (N.C.H.); (G.B.)
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Young A, Oldham E, Hittner M, Beerman S, Chancellor M, Smith CA, Nees D, Hughes G, Vassar M. Endorsement of reporting guidelines and clinical trial registration by tropical medicine and infectious disease journals: A cross-sectional study. Trop Med Int Health 2025. [PMID: 40365862 DOI: 10.1111/tmi.14127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
BACKGROUND Studies published in academic medical journals inform and influence healthcare decisions. Sufficient study reporting is primarily charged to researchers. However, journals can promote more complete reporting of their published studies. Recommending or requiring reporting guideline use and prospective trial registration may ensure published studies adhere to rigorous reporting standards. This study aimed to evaluate 'instructions to authors' pages of tropical medicine and infectious disease (TM/ID) journals to assess endorsement of reporting guidelines (RGs) for common medical study designs and clinical trial registration. METHODS Using a cross-sectional design guided by the Strengthening the Reporting of Observational Studies in Epidemiology checklist, we examined the top 100 infectious disease (ID) journals identified by the 2021 Scopus CiteScore tool and the 21 tropical medicine (TM) journals identified by Clarivate Web of Science. Each editorial journal staff was contacted for specific study designs accepted. Data were extracted from journals' 'instructions to authors' webpages with any discrepancies being resolved through consensus. We assessed adherence to RGs and clinical trial registration. RESULTS This study identified 293 TM/ID journals. Among the top 100 ID journals selected (Scopus CiteScore), 2 unfit journals were replaced. Among the 28 TM journals selected (Clarivate Web of Science), 5 were removed due to being duplicates and 2 were removed due to not being published in English. The Enhancing the QUAlity and Transparency Of health Research (EQUATOR) Network was cited by 49% of journals, while 85% of journals referenced the International Committee of Medical Journal Editors (ICMJE). Consolidated Standards of Reporting Trials (CONSORT) was most cited (73%), Quality of Reporting of Meta-analyses was least (2.6%). Clinical trial registration was mentioned by 73% of the journals. CONCLUSIONS TM and ID journals demonstrated suboptimal endorsement of various RGs. Among our findings, however, CONSORT and clinical trial registration garnered over 70% endorsement. We propose journals streamline RGs, establish user-friendly 'instructions to authors' pages and mandate reporting guideline adherence. These insights inform future research on enhancing reporting guideline use and TM/ID research quality.
Collapse
Affiliation(s)
- Alec Young
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Eli Oldham
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Makenna Hittner
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Skylarr Beerman
- New York Institute of Technology College of Osteopathic Medicine, Arkansas State University, Jonesboro, Arkansas, USA
| | - Matthew Chancellor
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Caleb A Smith
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Danya Nees
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Griffin Hughes
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Matt Vassar
- Office of Medical Student Research, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
- Department of Psychiatry and Behavioral Sciences, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA
| |
Collapse
|
4
|
Putz EJ, Andreasen CB, Stasko JA, Hamond C, Olsen SC, Nally JE, Palmer MV. Circulating foamy macrophages and other features of bacillus Calmette-Guérin challenge in Golden Syrian hamsters. Vaccine 2025; 55:127037. [PMID: 40117727 DOI: 10.1016/j.vaccine.2025.127037] [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: 04/30/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Golden Syrian Hamsters are utilized as rodent research models for various bacterial diseases. They are highly susceptible to leptospirosis, making hamsters the most common model for testing and maintaining virulence of laboratory Leptospira strains as well as for bacterin vaccine efficiency testing. Hamsters are also used for modeling features of Bacillus Calmette-Guérin (BCG) and tuberculosis, as they consistently develop granulomas, differing from other BCG animal models. Circulating foamy macrophages have recently been identified in the blood of the hamsters following Leptospira challenge but not in controls. It has been unknown whether this phenomenon is specific to Leptospira/hamster interactions, or whether hamsters will produce circulating foamy macrophages in response to other bacterial infections. In this study, we established that hamsters develop circulating foamy macrophages when challenged intraperitoneally with BCG or Leptospira. In addition to circulating foamy macrophages, hamsters infected with BCG had widespread granuloma formation in major organs and injection sites which also contained resident foamy macrophages, as well as mineralized bodies, sometimes containing acid fast bacteria.
Collapse
Affiliation(s)
- E J Putz
- Infectious Bacterial Disease Research Unit, USDA Agriculture Research Service, National Animal Disease Center, 1920 Dayton Ave, Ames, IA 50010, USA; Department of Veterinary Pathology, College of Veterinary Medicine, 1800 Christensen Drive, Ames, IA, 50010, USA.
| | - C B Andreasen
- Department of Veterinary Pathology, College of Veterinary Medicine, 1800 Christensen Drive, Ames, IA, 50010, USA
| | - J A Stasko
- Infectious Bacterial Disease Research Unit, USDA Agriculture Research Service, National Animal Disease Center, 1920 Dayton Ave, Ames, IA 50010, USA
| | - C Hamond
- National Veterinary Services Laboratories, APHIS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - S C Olsen
- Infectious Bacterial Disease Research Unit, USDA Agriculture Research Service, National Animal Disease Center, 1920 Dayton Ave, Ames, IA 50010, USA
| | - J E Nally
- Infectious Bacterial Disease Research Unit, USDA Agriculture Research Service, National Animal Disease Center, 1920 Dayton Ave, Ames, IA 50010, USA
| | - M V Palmer
- Infectious Bacterial Disease Research Unit, USDA Agriculture Research Service, National Animal Disease Center, 1920 Dayton Ave, Ames, IA 50010, USA
| |
Collapse
|
5
|
Wang YM, Wang WC, Pan Y, Zeng L, Wu J, Wang ZB, Zhuang XL, Li ML, Cooper DN, Wang S, Shao Y, Wang LM, Fan YY, He Y, Hu XT, Wu DD. Regional and aging-specific cellular architecture of non-human primate brains. Genome Med 2025; 17:41. [PMID: 40296047 PMCID: PMC12038948 DOI: 10.1186/s13073-025-01469-x] [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: 01/19/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Deciphering the functionality and dynamics of brain networks across different regions and age groups in non-human primates (NHPs) is crucial for understanding the evolution of human cognition as well as the processes underlying brain pathogenesis. However, systemic delineation of the cellular composition and molecular connections among multiple brain regions and their alterations induced by aging in NHPs remain largely unresolved. METHODS In this study, we performed single-nucleus RNA sequencing on 39 samples collected from 10 brain regions of two young and two aged rhesus macaques using the DNBelab C4 system. Validation of protein expression of signatures specific to particular cell types, brain regions, and aging was conducted through a series of immunofluorescence and immunohistochemistry staining experiments. Loss-of-function experiments mediated by short hairpin RNA (shRNA) targeting two age-related genes (i.e., VSNL1 and HPCAL4) were performed in U251 glioma cells to verify their aging effects. Senescence-associated beta-galactosidase (SA-β-gal) staining and quantitative PCR (qPCR) of senescence marker genes were employed to assess cellular senescence in U251 cells. RESULTS We have established a large-scale cell atlas encompassing over 330,000 cells for the rhesus macaque brain. Our analysis identified numerous gene expression signatures that were specific to particular cell types, subtypes, brain regions, and aging. These datasets greatly expand our knowledge of primate brain organization and highlight the potential involvement of specific molecular and cellular components in both the regionalization and functional integrity of the brain. Our analysis also disclosed extensive transcriptional alterations and cell-cell connections across brain regions in the aging macaques. Finally, by examining the heritability enrichment of human complex traits and diseases, we determined that neurological traits were significantly enriched in neuronal cells and multiple regions with aging-relevant gene expression signatures, while immune-related traits exhibited pronounced enrichment in microglia. CONCLUSIONS Taken together, our study presents a valuable resource for investigating the cellular and molecular architecture of the primate nervous system, thereby expanding our understanding of the mechanisms underlying brain function, aging, and disease.
Collapse
Affiliation(s)
- Yun-Mei Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wen-Chao Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yongzhang Pan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin Zeng
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jing Wu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Zheng-Bo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650107, China
| | - Xiao-Lin Zhuang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ming-Li Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sheng Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yong Shao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Li-Min Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Ying-Yin Fan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yonghan He
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xin-Tian Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.
| |
Collapse
|
6
|
Pimenta JC, Beltrami VA, Oliveira BDS, Queiroz-Junior CM, Barsalini J, Teixeira DC, de Souza-Costa LP, Lima ALD, Machado CA, Parreira BZSG, Santos FRDS, Costa PAC, Lacerda LDSB, Gonçalves MR, Chaves IDM, Couto MGG, Costa VRDM, Nóbrega NRC, Silva BL, Fonseca T, Resende F, Wnuk NT, Marim FM, Rocha FEO, Umezu HL, Campolina-Silva G, Andrade ACDSP, de Aguiar RS, Costa GMJ, Guimarães PPG, Silva GSF, Rachid MA, Vieira LB, Pinho V, Teixeira AL, Teixeira MM, Miranda AS, Costa VV. Neuropsychiatric sequelae in an experimental model of post-COVID syndrome in mice. Brain Behav Immun 2025; 128:16-36. [PMID: 40120834 DOI: 10.1016/j.bbi.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
The global impact of the COVID-19 pandemic has been unprecedented, and presently, the world is facing a new challenge known as post-COVID syndrome (PCS). Current estimates suggest that more than 100 million people are grappling with PCS, encompassing several manifestations, including pulmonary, musculoskeletal, metabolic, and neuropsychiatric sequelae (cognitive and behavioral). The mechanisms underlying PCS remain unclear. The present study aimed to: (i) comprehensively characterize the acute effects of pulmonary inoculation of the betacoronavirus MHV-A59 in immunocompetent mice at clinical, cellular, and molecular levels; (ii) examine potential acute and long-term pulmonary, musculoskeletal, and neuropsychiatric sequelae induced by the betacoronavirus MHV-A59; and to (iii) assess sex-specific differences. Male and female C57Bl/6 mice were initially inoculated with varying viral titers (3x103 to 3x105 PFU/30 μL) of the betacoronavirus MHV-A59 via the intranasal route to define the highest inoculum capable of inducing disease without causing mortality. Further experiments were conducted with the 3x104 PFU inoculum. Mice exhibited an altered neutrophil/lymphocyte ratio in the blood in the 2nd and 5th day post-infection (dpi). Marked lung lesions were characterized by hyperplasia of the alveolar walls, infiltration of polymorphonuclear leukocytes (PMN) and mononuclear leukocytes, hemorrhage, increased concentrations of CCL2, CCL3, CCL5, and CXCL1 chemokines, as well as high viral titers until the 5th dpi. While these lung inflammatory signs resolved, other manifestations were observed up to the 60 dpi, including mild brain lesions with gliosis and hyperemic blood vessels, neuromuscular dysfunctions, anhedonic-like behavior, deficits in spatial working memory, and short-term aversive memory. These musculoskeletal and neuropsychiatric complications were exclusive to female mice and prevented after ovariectomy. In summary, our study describes for the first time a novel sex-dependent model of PCS focused on neuropsychiatric and musculoskeletal disorders. This model provides a unique platform for future investigations regarding the effects of acute therapeutic interventions on the long-term sequelae unleashed by betacoronavirus infection.
Collapse
Affiliation(s)
- Jordane Clarisse Pimenta
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vinícius Amorim Beltrami
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruna da Silva Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jéssica Barsalini
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle Cunha Teixeira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz Pedro de Souza-Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anna Luiza Diniz Lima
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline Amaral Machado
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Felipe Rocha da Silva Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Matheus Rodrigues Gonçalves
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Manoela Gonzaga Gontijo Couto
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Rodrigues de Melo Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Bárbara Luísa Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Talita Fonseca
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Filipe Resende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Teixeira Wnuk
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe Emanuel Oliveira Rocha
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Hanna L Umezu
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabriel Campolina-Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Ana Cláudia Dos Santos Pereira Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Microbiology and Immunology, Université Laval, Quebec, Canada
| | - Renato Santana de Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme Mattos Jardim Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Glauber Santos Ferreira Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Alvarenga Rachid
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Biggs Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva Miranda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Ghare S, Warner D, Warner J, Chilton PM, Lee J, Zhang J, Wang M, Hardesty J, Treves R, Gabbard J, Anderson C, Batra L, Sreenivasan C, Kraenzle J, McCulley M, McCoy S, Zhang L, Feng W, Gondim DD, Barve S, Zheng J, Palmer K, McClain C, Kirpich I. Impact of chronic ethanol consumption and SARS-COV-2 on the liver and intestine: A pilot dose-response study in mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:587-598. [PMID: 39757351 PMCID: PMC11928281 DOI: 10.1111/acer.15528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND During the coronavirus disease 2019 (COVID-19) pandemic, there was a marked increase in alcohol consumption. COVID-19 superimposed on underlying liver disease notably worsens the outcome of many forms of liver injury. The goal of a current pilot study was to test the dual exposure of alcohol and COVID-19 infection in an experimental animal model of alcohol-associated liver disease (ALD). METHODS After 4 weeks of ethanol (EtOH) feeding, C57BL/6 male mice received SARS-CoV-2 (SARS2-N501YMA30) intranasally at 3 × 102, 1 × 103, 3 × 103, and 1 × 104 plaque-forming units (PFU). Mice were then weighed/monitored daily for morbidity/mortality for 10 days while continuing EtOH consumption. Markers of liver inflammation, injury, and intestinal barrier integrity were evaluated. RESULTS A similar gradual weight loss was observed in all inoculated mice (slightly less in the 3 × 102 group) up to post-infection day 4. Greater mortality was observed in mice receiving the highest viral dose at days 3 and 4 post-infection. The majority of the surviving mice subjected to EtOH and inoculated with 3 × 103 or 1 × 104 PFU rapidly lost 25% of their body weight and were euthanized on post-infection day 4. Analysis of liver health in animals that survived to the end of the experiment exhibited no significant changes in hepatic steatosis but had a limited increase in plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels at all viral doses versus EtOH alone. However, the 1 × 104 PFU viral dose exacerbated EtOH-induced hepatic inflammation characterized by elevated levels of several pro-inflammatory cytokines, including Il-6 and Tnf-α. There was limited effect of viral infection on the intestine. CONCLUSIONS SARS-CoV-2 infection caused a dose-dependent negative impact on body weight and survival in mice fed EtOH. This pilot study suggests that early mortality observed after high-dose SARS-CoV-2 challenge could be due, in part, to hepatic dysfunction following chronic EtOH feeding.
Collapse
Affiliation(s)
- Smita Ghare
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Dennis Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jeffrey Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Paula M. Chilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jiyeon Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - JingWen Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Min Wang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Josiah Hardesty
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Rui Treves
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jon Gabbard
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Charles Anderson
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Lalit Batra
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Chithra Sreenivasan
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jennifer Kraenzle
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Matthew McCulley
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Stephanie McCoy
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Lihua Zhang
- Department of Structural & Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, United States
| | - Wenke Feng
- Department of Structural & Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, United States
| | - Dibson Dibe Gondim
- Department of Pathology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Shirish Barve
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jian Zheng
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Kenneth Palmer
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Craig McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Robley Rex Veterans Affairs Medical Center, 800 Zorn Avenue, Louisville, KY 40206, United States
| | - Irina Kirpich
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| |
Collapse
|
8
|
Jiang RD, Luo YZ, Lin HF, Zheng XS, Zeng WT, Liu MQ, Deng HH, Wang Q, Lai YN, Chen Y, Guo ZS, Zeng Y, Gong QC, Qiu C, Dong M, Wang X, Wang ZY, Ji LN, Hou PP, Li Q, Shen XR, Li B, Gao Y, Zhang AH, Jiang TT, Shi AM, Zhou P, Lin XH, Deng ZQ, Li JM, Shi ZL. Impaired inflammatory resolution with severe SARS-CoV-2 infection in leptin knock out obese hamster. iScience 2025; 28:111837. [PMID: 39981511 PMCID: PMC11841202 DOI: 10.1016/j.isci.2025.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/28/2024] [Accepted: 12/13/2024] [Indexed: 02/22/2025] Open
Abstract
Comorbidities, such as obesity, increase the risk of severe COVID-19. However, the mechanisms underlying severe illnesses in individuals with obesity are poorly understood. Here, we used gene-edited leptin knock out (Leptin -/-) obese hamsters to establish a severe infection model. This model exhibits robust viral replication, severe lung lesions, pronounced clinical symptoms, and fatal infection, mirroring severe COVID-19 in patients with obesity. Using single-cell transcriptomics on lung tissues pre- and post-infection, we found that monocyte-derived alveolar macrophages (MD-AM) play a key role in lung hyper-inflammation, including two unique MD-AM cell fate branches specific to Leptin -/- hamsters. Notably, reduced Trem2-dependent efferocytosis pathways in Leptin -/- hamsters indicated weakened inflammation resolution, consistent with the scRNA-seq data from patients with obesity. In summary, our study highlights the obesity-associated mechanisms underlying severe SARS-CoV-2 infections and establishes a reliable preclinical animal model for developing obesity-specific therapeutics for critical COVID-19.
Collapse
Affiliation(s)
- Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Zhe Luo
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shuang Zheng
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hao-Hao Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Na Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zi-Shuo Guo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zeng
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Yi Wang
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Li-Na Ji
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Pan-Pan Hou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qian Li
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xu-Rui Shen
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Jiang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Zhou
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xin-Hua Lin
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Zi-Qing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
9
|
Zhang Y, Chen H, Li Y, Luo C, Zhu Y, Zhou X, Wang R, He J, Guo H, Xu X, Qiu M, Li J. Animal Models for Long COVID: Current Advances, Limitations, and Future Directions. J Med Virol 2025; 97:e70237. [PMID: 39981885 DOI: 10.1002/jmv.70237] [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: 10/20/2024] [Revised: 01/25/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
Long COVID (LC) represents a chronic, systemic, and often disabling condition that poses a significant ongoing threat to public health. Foundational scientific studies are needed to unravel the underlying mechanisms, with the ultimate goal of developing effective preventative and therapeutic strategies. Therefore, there is an urgent demand for animal models that can accurately replicate the clinical features of LC. This review integrates clinical epidemiological data to summarize the pathological changes in extrapulmonary systems involved in LC. Additionally, it critically examines the capacity of existing animal models, including nonhuman primates, genetically modified mice, and Syrian hamsters, to exhibit enduring postinfection symptoms that align with human clinical manifestations, and identifies key areas requiring further development. The objective is to offer insights that will aid in the development of next-generation animal models, thereby accelerating our understanding of how acute respiratory viral infections transition into chronic conditions, and ensuring preparedness for future pandemics.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Huan Chen
- Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Yumeng Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Chenxi Luo
- The Fifth Camp of Cadet Brigade, School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yunkai Zhu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiaoyang Zhou
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Ruixuan Wang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Jiuxiang He
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Hongxia Guo
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiaofeng Xu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Minyue Qiu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Jintao Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Liu M, Wang M, Sun X, Mu J, Teng T, Jin N, Song J, Li B, Zhang D. Polypropylene microplastics triggered mouse kidney lipidome reprogramming combined with ROS stress as revealed by lipidomics and Raman biospectra. CHEMOSPHERE 2025; 370:143926. [PMID: 39667527 DOI: 10.1016/j.chemosphere.2024.143926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Microplastics intrigue kidney toxicity such as mitochondrial dysfunction and inflammation promotion. However, as an organ relying heavily on fatty acid oxidation, how microplastics influence kidney lipidomes remain unclear. Hence, we performed Raman spectra and multidimensional mass spectrometry-based shotgun lipidomics to decode kidney lipidomics landscape under polypropylene microplastics exposure. Kidney functions and cellular redox homeostasis were remarkably disturbed as revealed by levels of biochemical renal function markers, malonaldehyde, hydrogen peroxide and antioxidants. Ultrastructure alterations including the foot process fusion implied the kidney injury associated with lipidomic changes. Raman spectra successfully further confirmed the cellular change of reactive oxygen species and lipid disorders. Lipidomics showed that polypropylene microplastics caused abnormal lipidome and irregular exchange by remodeling triglycerides and phospholipids. Genes involved in lipid metabolism such as Fads1 and Elovl5 exhibited highly diversified expression profiles responding to polypropylene microplastics stress and possessed significant correlations with ROS indicators. These results explained ultrastructure alterations and aggravation of kidney injuries. Our work revealed polypropylene microplastics inducing lipidomic detriment in mouse kidney by Raman spectra and lipidomics firstly, elucidating the significances of lipidomic remodeling coupled with ROS stress in the kidney damages. The findings provided reliable evidence on the health risks of polypropylene microplastics in kidney.
Collapse
Affiliation(s)
- Mingying Liu
- Key Labortary of Blood-stasis-toxin Syndrome of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Miao Wang
- Key Labortary of Blood-stasis-toxin Syndrome of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Xinglin Sun
- Key Labortary of Blood-stasis-toxin Syndrome of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Ju Mu
- Key Labortary of Blood-stasis-toxin Syndrome of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Tingting Teng
- Key Laboratory of Groundwater Resources and Environment, Ministry of Education, Changchun, 130021, PR China; College of New Energy and Environment, Jilin University, Changchun, 130021, PR China
| | - Naifu Jin
- College of Water Sciences, Beijing Normal University, Beijing, 100875, PR China
| | - Jiaxuan Song
- College of Water Sciences, Beijing Normal University, Beijing, 100875, PR China
| | - Bei Li
- State Key Lab of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; HOOKE Instruments Ltd., Changchun, 130033, PR China
| | - Dayi Zhang
- Key Laboratory of Groundwater Resources and Environment, Ministry of Education, Changchun, 130021, PR China; College of New Energy and Environment, Jilin University, Changchun, 130021, PR China; Key Laboratory of Regional Environment and Eco-restoration, Ministry of Education, Shenyang University, Shenyang, 110044, PR China.
| |
Collapse
|
11
|
Liu Y, Feng T, Tong W, Guo Z, Li X, Kong Q, Xiang Z. The transcriptome of MHV-infected RAW264.7 cells offers an alternative model for macrophage innate immunity research. Animal Model Exp Med 2025; 8:57-66. [PMID: 38992966 PMCID: PMC11798737 DOI: 10.1002/ame2.12443] [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: 01/14/2024] [Accepted: 04/15/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Macrophages are the primary innate immune cells encountered by the invading coronaviruses, and their abilities to initiate inflammatory reactions, to maintain the immunity homeostasis by differential polarization, to train the innate immune system by epigenic modification have been reported in laboratory animal research. METHODS In the current in vitro research, murine macrophage RAW 264.7 cell were infected by mouse hepatitis virus, a coronavirus existed in mouse. At 3-, 6-, 12-, 24-, and 48-h post infection (hpi.), the attached cells were washed with PBS and harvested in Trizol reagent. Then The harvest is subjected to transcriptome sequencing. RESULTS The transcriptome analysis showed the immediate (3 hpi.) up regulation of DEGs related to inflammation, like Il1b and Il6. DEGs related to M2 differential polarization, like Irf4 showed up regulation at 24 hpi., the late term after viral infection. In addition, DEGs related to metabolism and histone modification, like Ezh2 were detected, which might correlate with the trained immunity of macrophages. CONCLUSIONS The current in vitro viral infection study showed the key innated immunity character of macrophages, which suggested the replacement value of viral infection cells model, to reduce the animal usage in preclinical research.
Collapse
Affiliation(s)
- Yun Liu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Ting‐Ting Feng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Wei Tong
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Zhi Guo
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Xia Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Qi Kong
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| | - Zhi‐Guang Xiang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingP.R. China
| |
Collapse
|
12
|
Kenney D, O’Connell AK, Tseng AE, Turcinovic J, Sheehan ML, Nitido AD, Montanaro P, Gertje HP, Ericsson M, Connor JH, Vrbanac V, Crossland NA, Harly C, Balazs AB, Douam F. Immune Signatures of SARS-CoV-2 Infection Resolution in Human Lung Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583965. [PMID: 38496468 PMCID: PMC10942442 DOI: 10.1101/2024.03.08.583965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
While human autopsy samples have provided insights into pulmonary immune mechanisms associated with severe viral respiratory diseases, the mechanisms that contribute to a clinically favorable resolution of viral respiratory infections remain unclear due to the lack of proper experimental systems. Using mice co-engrafted with a genetically matched human immune system and fetal lung xenograft (fLX), we mapped the immunological events defining successful resolution of SARS-CoV-2 infection in human lung tissues. Viral infection is rapidly cleared from fLX following a peak of viral replication, histopathological manifestations of lung disease and loss of AT2 program, as reported in human COVID-19 patients. Infection resolution is associated with the activation of a limited number of hematopoietic subsets, including inflammatory monocytes and non-canonical double-negative T-cells with cytotoxic functions, which are highly enriched in viral RNA and dissipate upon infection resolution. Activation of specific human fibroblast and endothelial subsets also elicit robust antiviral and monocyte chemotaxis signatures, respectively. Notably, systemic depletion of human CD4+ cells, but not CD3+ cells, abrogates infection resolution in fLX and induces persistent infection, supporting evidence that peripheral CD4+ monocytes are important contributors to SARS-CoV-2 infection resolution in lung tissues. Collectively, our findings unravel a comprehensive picture of the immunological events defining effective resolution of SARS-CoV-2 infection in human lung tissues, revealing markedly divergent immunological trajectories between resolving and fatal COVID-19 cases.
Collapse
Affiliation(s)
- Devin Kenney
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Aoife K. O’Connell
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Anna E. Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jacquelyn Turcinovic
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Maegan L. Sheehan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Adam D. Nitido
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Paige Montanaro
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hans P. Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maria Ericsson
- Electron Microscopy Core Facility, Harvard Medical School, Boston, MA, USA
| | - John H. Connor
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | | | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Christelle Harly
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France
- LabEx IGO ‘Immunotherapy, Graft, Oncology’, Nantes, France
- These authors contributed equally to the work
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- These authors contributed equally to the work
- Lead contact
| |
Collapse
|
13
|
Luo EY, Chuen-Chung Chang R, Gilbert-Jaramillo J. SARS-CoV-2 infection in microglia and its sequelae: What do we know so far? Brain Behav Immun Health 2024; 42:100888. [PMID: 39881814 PMCID: PMC11776083 DOI: 10.1016/j.bbih.2024.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 01/31/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the COVID-19 pandemic. After the success of therapeutics and worldwide vaccination, the long-term sequelae of SARS-CoV-2 infections are yet to be determined. Common symptoms of COVID-19 include the loss of taste and smell, suggesting SARS-CoV-2 infection has a potentially detrimental effect on neurons within the olfactory/taste pathways, with direct access to the central nervous system (CNS). This could explain the detection of SARS-CoV-2 antigens in the brains of COVID-19 patients. Different viruses display neurotropism that causes impaired neurodevelopment and/or neurodegeneration. Hence, it is plausible that COVID-19-associated neuropathologies are directly driven by SARS-CoV-2 infection in the CNS. Microglia, resident immune cells of the brain, are constantly under investigation as their surveillance role has been suggested to act as a friend or a foe impacting the progression of neurological disorders. Herein, we review the current literature suggesting microglia potentially been a susceptible target by SARS-CoV-2 virions and their role in viral dissemination within the CNS. Particular attention is given to the different experimental models and their translational potential.
Collapse
Affiliation(s)
- Echo Yongqi Luo
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Javier Gilbert-Jaramillo
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| |
Collapse
|
14
|
Yuya W, Yuansong Y, Susu L, Chen L, Yong W, Yining W, YouChun W, Changfa F. Progress and challenges in development of animal models for dengue virus infection. Emerg Microbes Infect 2024; 13:2404159. [PMID: 39312399 PMCID: PMC11423536 DOI: 10.1080/22221751.2024.2404159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
ABSTRACTThe severity of the dengue epidemic is on the rise, with its geographic range had expanded to southern Europe by 2024. In this August, the WHO updated the pathogens that could spark the next pandemic, dengue virus was on the list. Vaccines and drugs serve as powerful tools for both preventing dengue infections and treating patients. Animal models play a pivotal role in vaccine development and drug screening. Available potential susceptible animals, including non-human primates, rodents, pigs, and tree shrews, have been extensively explored to establish animal models of dengue disease. Despite significant advancements, there are still notable limitations. Different animal models exhibit distinct constraining factors such as viraemia, host susceptibility, immune function of the host, clinical symptoms, ADE (antibody-dependent enhancement) phenomena, cytokine storm response to various serotypes and strain variations. Furthermore, despite extensive research on the dengue virus receptor in recent years, genetically modified animal models immunocompetent harbouring dengue virus susceptibility receptors have not yet been available. This work reviewed the research progress of dengue virus receptors and dengue animal models, suggesting that the development of genetically modified murine models expressing dengue virus functional receptors may hold a promise for future dengue disease research, especially for its vaccine development.
Collapse
Affiliation(s)
- Wang Yuya
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Yang Yuansong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Liu Susu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Ling Chen
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
- College of Life Science school, Northwest University, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Xi’an, People’s Republic of China
| | - Wu Yong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang Yining
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang YouChun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| | - Fan Changfa
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| |
Collapse
|
15
|
Zhang G, Zhao B, Liu J. The Development of Animal Models for Respiratory Syncytial Virus (RSV) Infection and Enhanced RSV Disease. Viruses 2024; 16:1701. [PMID: 39599816 PMCID: PMC11598872 DOI: 10.3390/v16111701] [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: 09/04/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The development of immunoprophylactic products against respiratory syncytial virus (RSV) has resulted in notable advancements, leading to an increased demand for preclinical experiments and placing greater demands on animal models. Nevertheless, the field of RSV research continues to face the challenge of a lack of ideal animal models. Despite the demonstration of efficacy in animal studies, numerous RSV vaccine candidates have been unsuccessful in clinical trials, primarily due to the lack of suitable animal models. The most commonly utilized animal models for RSV research are cotton rats, mice, lambs, and non-human primates. These animals have been extensively employed in mechanistic studies and in the development and evaluation of vaccines and therapeutics. However, each model only exemplifies some, but not all, aspects of human RSV disease. The aim of this study was to provide a comprehensive summary of the disease symptoms, viral replication, pathological damage, and enhanced RSV disease (ERD) conditions across different RSV animal models. Furthermore, the advantages and disadvantages of each model are discussed, with the intention of providing a valuable reference for related RSV research.
Collapse
Affiliation(s)
| | - Binbin Zhao
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China;
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China;
| |
Collapse
|
16
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
17
|
Vorobyeva DA, Potashnikova DM, Maryukhnich EV, Rusakovich GI, Tvorogova AV, Kalinskaya AI, Pinegina NV, Kovyrshina AV, Dolzhikova IV, Postnikov AB, Rozov FN, Sotnikova TN, Kanner DY, Logunov DY, Gintsburg AL, Vasilieva EJ, Margolis LB. Cytokine production in an ex vivo model of SARS-CoV-2 lung infection. Front Immunol 2024; 15:1448515. [PMID: 39497823 PMCID: PMC11532052 DOI: 10.3389/fimmu.2024.1448515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction The mechanisms of the SARS-CoV-2-triggered complex alterations in immune cell activation and production of cytokines in lung tissue remain poorly understood, in part because of the limited use of adequate tissue models that simulate the structure and cell composition of the lung in vivo. We developed a novel ex vivo model of SARS-CoV-2 infection of lung explants, that maintains the intact tissue composition and the viral load for up to 7-10 days. Using this model, we studied cytokine production during SARS-CoV-2 infection. Materials and methods Lung tissue was monitored for viability and cell composition using flow cytometry and histological analysis. SARS-CoV-2 infection was verified immunohistochemically, viral loads in tissue and culture medium were monitored by qPCR. A panel of 41 cytokines was measured in culture medium using xMAP technology. Results The explant lung tissue was viable and maintained viral infection that influenced the cytokine production. Elevated concentrations of G-CSF, GM-CSF, GRO-a, IFN-g, IL-6, IL-8, IP-10, MCP-3, MIP-1a, PDGF-AA, and VEGF, and decreased IL-1RA concentration were observed in infected tissue compared to non-infected tissue. Discussion Our results generally reflect the data obtained in COVID-19 patients. GRO-a, IFN-g, IL-6, IL-8, MCP-1, MCP-3, and RANTES correlated with the viral load, forming a distinct pro-inflammatory cluster. Thus, our lung ex vivo model faithfully reproduces some aspects of cytokine alterations in COVID-19 patients at an early disease stage, making the investigation of SARS-CoV-2 infection mechanisms more accessible and providing a potential platform for antiviral drug testing.
Collapse
Affiliation(s)
- Daria A. Vorobyeva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Daria M. Potashnikova
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Elena V. Maryukhnich
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - George I. Rusakovich
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Tvorogova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna I. Kalinskaya
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Natalia V. Pinegina
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Kovyrshina
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Inna V. Dolzhikova
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Tatiana N. Sotnikova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | | | - Denis Yu. Logunov
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L. Gintsburg
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena J. Vasilieva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Leonid B. Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
18
|
Jiang Q, Wu X, Dong F, Qiao S, Shi Q, Jian C, Chen C, Zhou J, Wang Y, Huang W. Construction of pseudotyped human coronaviruses and detection of pre-existing antibodies in the human population. BIOSAFETY AND HEALTH 2024; 6:279-285. [PMID: 40078732 PMCID: PMC11894997 DOI: 10.1016/j.bsheal.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 03/14/2025] Open
Abstract
In order to clarify the pre-exist immunity background of different human coronaviruses (HCoV), this study investigated the positive rate of spike (S) protein antibodies of HCoV, including HCoV- severe acute respiratory syndrome (SARS) -associated coronavirus (SARS-CoV-1), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Middle East respiratory syndrome coronavirus (MERS-CoV), HCoV-229E, HCoV-NL63, HCoV-HKU1 and HCoV-OC43, before and after the Coronavirus Disease 2019 (COVID-19) outbreak. We utilized pseudotyped virus-based neutralization assays (PBNA) or enzyme-linked immunosorbent assays (ELISA) to detect antibody levels against HCoV in serum samples collected in 2009-2010 and 2023. The PBNA results showed that neutralizing antibodies against SARS-CoV-1 and the MERS-CoV were negative. In the serum samples from 2009 to 2010, neutralizing antibodies against SARS-CoV-2 (D614G) were negative, whereas in the serum samples from 2023, 73 samples (73 %) showed neutralizing reactions with the SARS-CoV-2 D614G strain, 96 samples (96 %) with the BA.5 strain, and 91 samples (91 %) with the BF.7 strain. Among pre-COVID-19 samples, 33 % (33/100) showed neutralizing reactions with HCoV-229E and 63 % (63/100) with HCoV-NL63. Among post-COVID-19 samples, 50 % (50/100) showed neutralizing reactions with HCoV-229E and 49 % (49/100) with HCoV-NL63. Due to the different receptors of alpha coronavirus genus compared to other beta coronavirus genus, neutralizing antibodies against HCoV-OC43 and HCoV-HKU1 virus cannot be detected by constructing corresponding pseudotyped virus. Binding antibodies against HCoV-OC43 and HCoV-HKU1 virus were detected using ELISA. The results revealed that among pre-COVID-19 samples, 83 % (83/100) and 45 % (45/100) had binding activity with HCoV-OC43 and HCoV-HKU1, respectively. Among post-COVID-19 samples, 100 % (100/100) and 81 % (81/100) had binding activity with HCoV-OC43 and HCoV-HKU1, respectively.
Collapse
Affiliation(s)
- Qi Jiang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), State Key Laboratory of Drug Regulatory Science, Beijing 102629, China
| | - Xi Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), State Key Laboratory of Drug Regulatory Science, Beijing 102629, China
| | - Fangyu Dong
- Department of Research & Development, Taibang Biologic Group, Beijing 100125, China
| | - Shan Qiao
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing 102206, China
| | - Qiaoyun Shi
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing 102206, China
| | - Changyong Jian
- Department of Research & Development, Taibang Biologic Group, Beijing 100125, China
| | - Chen Chen
- Department of Research & Development, Taibang Biologic Group, Beijing 100125, China
| | - Jiuyue Zhou
- Department of Research & Development, Taibang Biologic Group, Beijing 100125, China
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), State Key Laboratory of Drug Regulatory Science, Beijing 102629, China
| |
Collapse
|
19
|
Friedrich VD, Pennitz P, Wyler E, Adler JM, Postmus D, Müller K, Teixeira Alves LG, Prigann J, Pott F, Vladimirova D, Hoefler T, Goekeri C, Landthaler M, Goffinet C, Saliba AE, Scholz M, Witzenrath M, Trimpert J, Kirsten H, Nouailles G. Neural network-assisted humanisation of COVID-19 hamster transcriptomic data reveals matching severity states in human disease. EBioMedicine 2024; 108:105312. [PMID: 39317638 DOI: 10.1016/j.ebiom.2024.105312] [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: 04/05/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Translating findings from animal models to human disease is essential for dissecting disease mechanisms, developing and testing precise therapeutic strategies. The coronavirus disease 2019 (COVID-19) pandemic has highlighted this need, particularly for models showing disease severity-dependent immune responses. METHODS Single-cell transcriptomics (scRNAseq) is well poised to reveal similarities and differences between species at the molecular and cellular level with unprecedented resolution. However, computational methods enabling detailed matching are still scarce. Here, we provide a structured scRNAseq-based approach that we applied to scRNAseq from blood leukocytes originating from humans and hamsters affected with moderate or severe COVID-19. FINDINGS Integration of data from patients with COVID-19 with two hamster models that develop moderate (Syrian hamster, Mesocricetus auratus) or severe (Roborovski hamster, Phodopus roborovskii) disease revealed that most cellular states are shared across species. A neural network-based analysis using variational autoencoders quantified the overall transcriptomic similarity across species and severity levels, showing highest similarity between neutrophils of Roborovski hamsters and patients with severe COVID-19, while Syrian hamsters better matched patients with moderate disease, particularly in classical monocytes. We further used transcriptome-wide differential expression analysis to identify which disease stages and cell types display strongest transcriptional changes. INTERPRETATION Consistently, hamsters' response to COVID-19 was most similar to humans in monocytes and neutrophils. Disease-linked pathways found in all species specifically related to interferon response or inhibition of viral replication. Analysis of candidate genes and signatures supported the results. Our structured neural network-supported workflow could be applied to other diseases, allowing better identification of suitable animal models with similar pathomechanisms across species. FUNDING This work was supported by German Federal Ministry of Education and Research, (BMBF) grant IDs: 01ZX1304B, 01ZX1604B, 01ZX1906A, 01ZX1906B, 01KI2124, 01IS18026B and German Research Foundation (DFG) grant IDs: 14933180, 431232613.
Collapse
Affiliation(s)
- Vincent D Friedrich
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany; Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Leipzig, Germany
| | - Peter Pennitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Emanuel Wyler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Julia M Adler
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Dylan Postmus
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Liverpool School of Tropical Medicine, Department of Tropical Disease Biology, Liverpool, United Kingdom
| | - Kristina Müller
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany
| | - Luiz Gustavo Teixeira Alves
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Julia Prigann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Gladstone Institutes, San Francisco, USA
| | - Fabian Pott
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Thomas Hoefler
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Cengiz Goekeri
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany; Cyprus International University, Faculty of Medicine, Nicosia, Cyprus
| | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany; Humboldt-Universität zu Berlin, Institut fuer Biologie, Berlin, Germany
| | - Christine Goffinet
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Liverpool School of Tropical Medicine, Department of Tropical Disease Biology, Liverpool, United Kingdom
| | - Antoine-Emmanuel Saliba
- Faculty of Medicine, Institute of Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany
| | - Markus Scholz
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany; University of Leipzig, Faculty of Mathematics and Computer Science, Leipzig, Germany
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Jakob Trimpert
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Holger Kirsten
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany.
| | - Geraldine Nouailles
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany.
| |
Collapse
|
20
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
21
|
Gordhan BG, Liebenberg D, Scarlatti G, Herrera C, Chiodi F, Martinson N, Fox J, Kana BD. Ex vivo challenge models for infectious diseases. Crit Rev Microbiol 2024; 50:785-804. [PMID: 37909097 DOI: 10.1080/1040841x.2023.2274855] [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: 02/03/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
Traditionally, molecular mechanisms of pathogenesis for infectious agents were studied in cell culture or animal models but have limitations on the extent to which the resulting data reflect natural infection in humans. The COVID-19 pandemic has highlighted the urgent need to rapidly develop laboratory models that enable the study of host-pathogen interactions, particularly the relative efficacy of preventive measures. Recently, human and animal ex vivo tissue challenge models have emerged as a promising avenue to study immune responses, screen potential therapies and triage vaccine candidates. This approach offers the opportunity to closely approximate human disease from the perspective of pathology and immune response. It has advantages compared to animal models which are expensive, lengthy and often require containment facilities. Herein, we summarize some recent advances in the development of ex vivo tissue challenge models for COVID-19, HIV-1 and other pathogens. We focus on the contribution of these models to enhancing knowledge of host-pathogen interactions, immune modulation, and their value in testing therapeutic agents. We further highlight the advantages and limitations of using ex vivo challenge models and briefly summarize how the use of organoids provides a useful advancement over current approaches. Collectively, these developments have enormous potential for the study of infectious diseases.
Collapse
Affiliation(s)
- Bhavna Gowan Gordhan
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Dale Liebenberg
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Carolina Herrera
- Department of Infectious Disease, Imperial College London, London, UK
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Neil Martinson
- Perinatal HIV Research Unit (PHRU), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Julie Fox
- Guys and St. Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Bavesh Davandra Kana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
22
|
Park U, Lee JH, Kim U, Jeon K, Kim Y, Kim H, Kang JI, Park MY, Park SH, Cha JS, Yoon GY, Jeong DE, Kim T, Oh S, Yoon SH, Jin L, Ahn Y, Lim MY, Han SR, Kim HY, Kim MH, Zhang YH, Kang JG, Lee MS, Jeon YK, Cho HS, Lee HW, Cho NH. A humanized ACE2 mouse model recapitulating age- and sex-dependent immunopathogenesis of COVID-19. J Med Virol 2024; 96:e29915. [PMID: 39279412 DOI: 10.1002/jmv.29915] [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: 04/15/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/18/2024]
Abstract
In the ongoing battle against coronavirus disease 2019 (COVID-19), understanding its pathogenesis and developing effective treatments remain critical challenges. The creation of animal models that closely replicate human infection stands as a critical step forward in this research. Here, we present a genetically engineered mouse model with specifically-humanized knock-in ACE2 (hiACE2) receptors. This model, featuring nine specific amino acid substitutions for enhanced interaction with the viral spike protein, enables efficient severe acute respiratory syndrome coronavirus 2 replication in respiratory organs without detectable infection in the central nervous system. Moreover, it mirrors the age- and sex-specific patterns of morbidity and mortality, as well as the immunopathological features observed in human COVID-19 cases. Our findings further demonstrate that the depletion of eosinophils significantly reduces morbidity and mortality, depending on the infecting viral dose and the sex of the host. This reduction is potentially achieved by decreasing the pathogenic contribution of eosinophil-mediated inflammation, which is strongly correlated with neutrophil activity in human patients. This underscores the model's utility in studying the immunopathological aspects of COVID-19 and represents a significant advancement in COVID-19 modeling. It offers a valuable tool for testing vaccines and therapeutics, enhancing our understanding of the disease mechanisms and potentially guiding more targeted and effective treatments.
Collapse
Affiliation(s)
- Uni Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
- GEMCRO Inc., Seoul, South Korea
| | - Uijin Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Kyeongseok Jeon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
| | - Hyeran Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Ju-Il Kang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
| | | | | | - Jeong Seok Cha
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Ga-Yeon Yoon
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Da-Eun Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do, South Korea
| | - Taehun Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Songhyeok Oh
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Ho Yoon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Physiology & Biomedical Sciences, Ischemic/hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Liyuan Jin
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoojin Ahn
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
| | - Min Yeong Lim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
| | - Seung Ro Han
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Hye Young Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
| | - Myoung-Hwan Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Physiology & Biomedical Sciences, Ischemic/hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
- Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
| | - Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, Ischemic/hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jun-Gu Kang
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do, South Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
- GEMCRO Inc., Seoul, South Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
- Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
23
|
Gorskaya AV, Vasilev DS. Problems in the Diagnosis of Dysfunctions of the Olfactory Analyzer in Laboratory Animals Based on Behavioral and Electrophysiological Study Methods. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2024; 54:990-1002. [DOI: 10.1007/s11055-024-01702-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2025]
|
24
|
Compeer B, Neijzen TR, van Lelyveld SFL, Martina BEE, Russell CA, Goeijenbier M. Uncovering the Contrasts and Connections in PASC: Viral Load and Cytokine Signatures in Acute COVID-19 versus Post-Acute Sequelae of SARS-CoV-2 (PASC). Biomedicines 2024; 12:1941. [PMID: 39335455 PMCID: PMC11428903 DOI: 10.3390/biomedicines12091941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
The recent global COVID-19 pandemic has had a profound and enduring impact, resulting in substantial loss of life. The scientific community has responded unprecedentedly by investigating various aspects of the crisis, particularly focusing on the acute phase of COVID-19. The roles of the viral load, cytokines, and chemokines during the acute phase and in the context of patients who experienced enduring symptoms upon infection, so called Post-Acute Sequelae of COVID-19 or PASC, have been studied extensively. Here, in this review, we offer a virologist's perspective on PASC, highlighting the dynamics of SARS-CoV-2 viral loads, cytokines, and chemokines in different organs of patients across the full clinical spectrum of acute-phase disease. We underline that the probability of severe or critical disease progression correlates with increased viral load levels detected in the upper respiratory tract (URT), lower respiratory tract (LRT), and plasma. Acute-phase viremia is a clear, although not unambiguous, predictor of PASC development. Moreover, both the quantity and diversity of functions of cytokines and chemokines increase with acute-phase disease severity. Specific cytokines remain or become elevated in the PASC phase, although the driving factor of ongoing inflammation found in patients with PASC remains to be investigated. The key findings highlighted in this review contribute to a further understanding of PASC and their differences and overlap with acute disease.
Collapse
Affiliation(s)
- Brandon Compeer
- Artemis Bioservices B.V., 2629 JD Delft, The Netherlands
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Tobias R Neijzen
- Department of Intensive Care Medicine, Spaarne Gasthuis, 2035 RC Haarlem, The Netherlands
| | | | | | - Colin A Russell
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Marco Goeijenbier
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
- Department of Intensive Care, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
25
|
Kar M, Johnson KEE, Vanderheiden A, Elrod EJ, Floyd K, Geerling E, Stone ET, Salinas E, Banakis S, Wang W, Sathish S, Shrihari S, Davis-Gardner ME, Kohlmeier J, Pinto A, Klein R, Grakoui A, Ghedin E, Suthar MS. CD4 + and CD8 + T cells are required to prevent SARS-CoV-2 persistence in the nasal compartment. SCIENCE ADVANCES 2024; 10:eadp2636. [PMID: 39178263 PMCID: PMC11343035 DOI: 10.1126/sciadv.adp2636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/19/2024] [Indexed: 08/25/2024]
Abstract
SARS-CoV-2 infection induces the generation of virus-specific CD4+ and CD8+ effector and memory T cells. However, the contribution of T cells in controlling SARS-CoV-2 during infection is not well understood. Following infection of C57BL/6 mice, SARS-CoV-2-specific CD4+ and CD8+ T cells are recruited to the respiratory tract, and a vast proportion secrete the cytotoxic molecule granzyme B. Using depleting antibodies, we found that T cells within the lungs play a minimal role in viral control, and viral clearance occurs in the absence of both CD4+ and CD8+ T cells through 28 days postinfection. In the nasal compartment, depletion of both CD4+ and CD8+ T cells, but not individually, results in persistent, culturable virus replicating in the nasal epithelial layer through 28 days postinfection. Viral sequencing analysis revealed adapted mutations across the SARS-CoV-2 genome, including a large deletion in ORF6. Overall, our findings highlight the importance of T cells in controlling virus replication within the respiratory tract during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Meenakshi Kar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Katherine E. E. Johnson
- Systems Genomics Section, Laboratory of Parasitic Diseases, DIR, NIAID, NIH, Bethesda, MD, USA
| | - Abigail Vanderheiden
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth J. Elrod
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Katharine Floyd
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Eduardo Salinas
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Stephanie Banakis
- Systems Genomics Section, Laboratory of Parasitic Diseases, DIR, NIAID, NIH, Bethesda, MD, USA
| | - Wei Wang
- Systems Genomics Section, Laboratory of Parasitic Diseases, DIR, NIAID, NIH, Bethesda, MD, USA
| | - Shruti Sathish
- Systems Genomics Section, Laboratory of Parasitic Diseases, DIR, NIAID, NIH, Bethesda, MD, USA
| | - Swathi Shrihari
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Meredith E. Davis-Gardner
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Jacob Kohlmeier
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Amelia Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Robyn Klein
- Schulich School of Medicine and Dentistry, Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Schulich School of Medicine and Dentistry, Western Institute of Neuroscience, Western University, London, Ontario, Canada
| | - Arash Grakoui
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, DIR, NIAID, NIH, Bethesda, MD, USA
| | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| |
Collapse
|
26
|
Jenkins F, Mapulanga T, Thapa G, da Costa KAS, Temperton NJ. Conference Report: LPMHealthcare Emerging Viruses 2023 (EVOX23): Pandemics-Learning from the Past and Present to Prepare for the Future. Pathogens 2024; 13:679. [PMID: 39204279 PMCID: PMC11357271 DOI: 10.3390/pathogens13080679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
The emergence of SARS-CoV-2 has meant that pandemic preparedness has become a major focus of the global scientific community. Gathered in the historic St Edmund Hall college in Oxford, the one-day LPMHealthcare conference on emerging viruses (6 September 2023) sought to review and learn from past pandemics-the current SARS-CoV-2 pandemic and the Mpox outbreak-and then look towards potential future pandemics. This includes an emphasis on monitoring the "traditional" reservoirs of viruses with zoonotic potential, as well as possible new sources of spillover events, e.g., bats, which we are coming into closer contact with due to climate change and the impacts of human activities on habitats. Continued vigilance and investment into creative scientific solutions is required for issues including the long-term physical and psychological effects of COVID-19, i.e., long COVID. The evaluation of current systems, including environmental monitoring, communication (with the public, regulatory authorities, and governments), and training; assessment of the effectiveness of the technologies/assays we have in place currently; and lobbying of the government and the public to work with scientists are all required in order to build trust moving forward. Overall, the SARS-CoV-2 pandemic has shown how many sectors can work together to achieve a global impact in times of crisis.
Collapse
Affiliation(s)
| | - Tobias Mapulanga
- Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham ME4 4BF, UK; (T.M.); (G.T.)
| | - Gauri Thapa
- Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham ME4 4BF, UK; (T.M.); (G.T.)
| | - Kelly A. S. da Costa
- Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham ME4 4BF, UK; (T.M.); (G.T.)
| | - Nigel J. Temperton
- Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham ME4 4BF, UK; (T.M.); (G.T.)
| |
Collapse
|
27
|
Guo S, Yan T, Gao M, Zhou Y, Zhang Z, Liu Y, Zhang G, Zhu Z, Fan C. Study on the susceptibility of bovine coronavirus to BALB/c mice. Microb Pathog 2024; 192:106717. [PMID: 38806136 DOI: 10.1016/j.micpath.2024.106717] [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: 02/25/2024] [Revised: 05/14/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
There are no other bovine coronavirus (BCoV) infection models except calves, which makes efficacy evaluation of vaccines and pathogenic mechanism research of BCoV inconvenient owing to their high value and inconvenient operation. This study aimed to establish a mouse model of BCoV infection. BCoV was used to infect 4-week-old male BALB/c mice and the optimal infection conditions were screened, including the following infection routes: gavage, intraperitoneal injection, and tail vein injection at doses of 1 × 108 TCID50, 2 × 108 TCID50 and 4 × 108 TCID50. Using the optimal infection conditions, BALB/c mice were infected with BCoV, and their body weight, blood routine, inflammatory factors, autopsy, virus distribution, and viral load were measured at 1, 3, 5, and 7 days after infection. The results showed that the optimal conditions for infecting BALB/c mice with BCoV HLJ-325 strain were continuous oral gavage for 3 days with a dose of 4 × 108 TCID50. On the 7th day after infection, there was significant extensive consolidation of the lungs and thinning of the colon wall. Significant inflammation was observed in various organs, especially in the colon and alveoli, where a large number of inflammatory cells infiltrate. Both BCoV Ag and nucleic acid are positive in visceral organs. The viral load in the colon and lungs was significantly higher than that in the other organs (p < 0.001). BCoV-infected mice showed a decreasing trend in body weight starting from day 5, and there was a significant difference compared to the control group on days 6 and 7 (p < 0.001). The total number of white blood cells and lymphocytes began to decrease and was significantly lower than that in the control group 24 h after infection (p < 0.001), and gradually returned to the control level. The cytokine TNF-α, IL-1β, and IL-6 showed an increasing trend, significantly higher than the control group on day 5 and 7 (p < 0.001). These results indicate that the BCoV HLJ-325 strain can infect BALB/c mice and cause inflammatory reactions and tissue lesions. The most significant effect was observed on the seventh day after infection with a dose of 4 × 108 TCID50 and three consecutive gavages. This study established, for the first time, a BALB/c mouse model of BCoV infection, providing a technical means for evaluating the immune efficacy of BCoV vaccines and studying their pathogenic mechanisms.
Collapse
Affiliation(s)
- Song Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Tingfu Yan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Mengmeng Gao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, China.
| | - Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Guohua Zhang
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, 161000, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Chunling Fan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| |
Collapse
|
28
|
Peidli S, Nouailles G, Wyler E, Adler JM, Kunder S, Voß A, Kazmierski J, Pott F, Pennitz P, Postmus D, Teixeira Alves LG, Goffinet C, Gruber AD, Blüthgen N, Witzenrath M, Trimpert J, Landthaler M, Praktiknjo SD. Single-cell-resolved interspecies comparison shows a shared inflammatory axis and a dominant neutrophil-endothelial program in severe COVID-19. Cell Rep 2024; 43:114328. [PMID: 38861386 DOI: 10.1016/j.celrep.2024.114328] [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: 12/15/2023] [Revised: 04/21/2024] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
A key issue for research on COVID-19 pathogenesis is the lack of biopsies from patients and of samples at the onset of infection. To overcome these hurdles, hamsters were shown to be useful models for studying this disease. Here, we further leverage the model to molecularly survey the disease progression from time-resolved single-cell RNA sequencing data collected from healthy and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected Syrian and Roborovski hamster lungs. We compare our data to human COVID-19 studies, including bronchoalveolar lavage, nasal swab, and postmortem lung tissue, and identify a shared axis of inflammation dominated by macrophages, neutrophils, and endothelial cells, which we show to be transient in Syrian and terminal in Roborovski hamsters. Our data suggest that, following SARS-CoV-2 infection, commitment to a type 1- or type 3-biased immunity determines moderate versus severe COVID-19 outcomes, respectively.
Collapse
Affiliation(s)
- Stefan Peidli
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany; Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Geraldine Nouailles
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Julia M Adler
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Sandra Kunder
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Anne Voß
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Julia Kazmierski
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Pott
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Pennitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Dylan Postmus
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Christine Goffinet
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Nils Blüthgen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany; Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Jakob Trimpert
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany; Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Markus Landthaler
- Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Samantha D Praktiknjo
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
29
|
Rochowski MT, Jayathilake K, Balcerak JM, Tamil Selvan M, Gunasekara S, Rudd J, Miller C, Lacombe VA. Alterations of whole body glucose metabolism in a feline SARS-CoV-2 infection model. Am J Physiol Regul Integr Comp Physiol 2024; 326:R499-R506. [PMID: 38574344 PMCID: PMC11381005 DOI: 10.1152/ajpregu.00228.2023] [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: 10/04/2023] [Revised: 03/06/2024] [Accepted: 03/30/2024] [Indexed: 04/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been especially devastating to patients with comorbidities, including metabolic and cardiovascular diseases. Elevated blood glucose during SARS-CoV-2 infection increased mortality of patients with COVID-19, although the mechanisms are not well understood. It has been previously demonstrated that glucose transport and utilization is a crucial pathway for other highly infectious RNA viruses. Thus, we hypothesized that SARS-CoV-2 infection could lead to alterations in cellular and whole body glucose metabolism. Specific pathogen-free domestic cats were intratracheally inoculated with USA-WA1/2020 (wild-type) SARS-CoV-2 or vehicle-inoculated, then euthanized at 4- and 8-days postinoculation (dpi). Blood glucose and cortisol concentrations were elevated at 4 and 8 dpi. Blood ketones, insulin, and angiotensin II concentrations remained unchanged throughout the experimental timeline. SARS-CoV-2 RNA was detected in the lung and heart, without changes in angiotensin-converting enzyme 2 (ACE2) RNA expression. In the lung, SARS-CoV-2 infection increased glucose transporter 1 (GLUT1) protein levels at 4 and 8 dpi, whereas GLUT4 level was only upregulated at 8 dpi. In the heart, GLUT-1 and -4 protein levels remained unchanged. Furthermore, GLUT1 level was upregulated in the skeletal muscle at 8 dpi, and AMPK was activated in the hearts of infected cats. SARS-CoV-2 infection increased blood glucose concentration and pulmonary GLUT protein levels. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming primarily in the lung to support viral replication. Furthermore, this translational feline model mimicked human COVID-19 and could be used to explore novel therapeutic targets to treat metabolic disease during SARS-CoV-2 infection.NEW & NOTEWORTHY Our study on a feline model of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, mirroring human COVID-19, revealed alterations in whole body and cellular glucose metabolism. Infected cats developed mild hyperglycemia, increased protein levels of glucose transporters in the lung, and AMPK activation in the heart. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming in the cardiorespiratory system to support viral replication. Understanding these mechanisms could lead to novel antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Matthew T Rochowski
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| | - Kaushalya Jayathilake
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - John-Michael Balcerak
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Miruthula Tamil Selvan
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Sachithra Gunasekara
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Jennifer Rudd
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Craig Miller
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Véronique A Lacombe
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
30
|
Qiu B, Zandkarimi F, Saqi A, Castagna C, Tan H, Sekulic M, Miorin L, Hibshoosh H, Toyokuni S, Uchida K, Stockwell BR. Fatal COVID-19 pulmonary disease involves ferroptosis. Nat Commun 2024; 15:3816. [PMID: 38769293 PMCID: PMC11106344 DOI: 10.1038/s41467-024-48055-0] [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: 08/18/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
SARS-CoV-2 infection causes severe pulmonary manifestations, with poorly understood mechanisms and limited treatment options. Hyperferritinemia and disrupted lung iron homeostasis in COVID-19 patients imply that ferroptosis, an iron-dependent cell death, may occur. Immunostaining and lipidomic analysis in COVID-19 lung autopsies reveal increases in ferroptosis markers, including transferrin receptor 1 and malondialdehyde accumulation in fatal cases. COVID-19 lungs display dysregulation of lipids involved in metabolism and ferroptosis. We find increased ferritin light chain associated with severe COVID-19 lung pathology. Iron overload promotes ferroptosis in both primary cells and cancerous lung epithelial cells. In addition, ferroptosis markers strongly correlate with lung injury severity in a COVID-19 lung disease model using male Syrian hamsters. These results reveal a role for ferroptosis in COVID-19 pulmonary disease; pharmacological ferroptosis inhibition may serve as an adjuvant therapy to prevent lung damage during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Baiyu Qiu
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Fereshteh Zandkarimi
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
- Mass Spectrometry Core Facility, Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Candace Castagna
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hui Tan
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Miroslav Sekulic
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-Cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Koji Uchida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, 10027, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
31
|
Li S, Lu Y, Yang S, Wang C, Yang J, Huang X, Chen G, Shao Y, Li M, Yu H, Fu Y, Liu G. Porcine lung tissue slices: a culture model for PRCV infection and innate immune response investigations. AMB Express 2024; 14:57. [PMID: 38753111 PMCID: PMC11098997 DOI: 10.1186/s13568-024-01717-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024] Open
Abstract
Respiratory coronaviruses (RCoVs) significantly threaten human health, necessitating the development of an ex vivo respiratory culture system for investigating RCoVs infection. Here, we successfully generated a porcine precision-cut lung slices (PCLSs) culture system, containing all resident lung cell types in their natural arrangement. Next, this culture system was inoculated with a porcine respiratory coronavirus (PRCV), exhibiting clinical features akin to humans who were infected by SARS-CoV-2. The results demonstrated that PRCV efficiently infected and replicated within PCLSs, targeting ciliated cells in the bronchioles, terminal bronchioles, respiratory bronchioles, and pulmonary alveoli. Additionally, through RNA-Seq analysis of the innate immune response in PCLSs following PRCV infection, expression levels of interferons, inflammatory cytokines and IFN stimulated genes were significantly upregulated. This ex vivo model may not only offer new insights into PRCV infection in the porcine respiratory tract but also serve as a valuable tool for studying human respiratory CoVs infection.
Collapse
Affiliation(s)
- Shuxian Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yabin Lu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Shanshan Yang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Caiying Wang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Jing Yang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Xin Huang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Guohui Chen
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Yongheng Shao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Maolin Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Haoyuan Yu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China
| | - Yuguang Fu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China.
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 XuJiaPing, YanChangBu, ChengGuan District, 730046, Lanzhou, Gansu, China.
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| |
Collapse
|
32
|
Wu J, Zhou Q, Qi H, Lan W, Yang S, Yang S, Fan Z, Zhang A. Antimicrobial resistance spectrum and virulence characterization of Escherichia coli, Klebsiella pneumoniae and Proteus mirabilis isolated from asymptomatic and diarrheal rhesus monkeys. Microbiol Res 2024; 282:127633. [PMID: 38364524 DOI: 10.1016/j.micres.2024.127633] [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: 10/11/2023] [Revised: 11/13/2023] [Accepted: 01/29/2024] [Indexed: 02/18/2024]
Abstract
This study aims to deepen our understanding of the drug resistance and virulence characterization among gut bacteria in asymptomatic and diarrheal captive rhesus macaques (RMs). A total of 31 samples, including 8 asymptomatic RMs, 10 diarrheal RMs, and 1 dead RM, were collected from a breeding base in Sichuan, China, for bacterial isolation. As a result, Escherichia coli (n = 23), Klebsiella (n = 22), Proteus mirabilis (n = 10), Enterococcus (n = 10), Salmonella (n = 2), and Staphylococcus (n = 2) were isolated. All isolates were subjected to antimicrobial susceptibility testing and whole-genome sequencing, among which some E. coli, K. pneumoniae, and P. mirabilis were subjected to the Galleria mellonella and mice infection testing. The antimicrobial resistance rates of levofloxacin, enrofloxacin, and cefotaxime in diarrhea-associated isolates were higher than those of asymptomatic isolates. Consistent with the antimicrobial resistance phenotype, diarrheal isolates had a higher prevalence rate to qnrS1, blaTEM-1B and blaCTX-M-27 than asymptomatic isolates. Furthermore, compared with asymptomatic isolates, diarrheal isolates demonstrated a higher pathogenic potential against larvae and mice. Additionally, sequence types (STs) 14179-14181 in E. coli and ST 625 and ST 630-631 in Klebsiella aerogenes were firstly characterized. Our evidence underscores the considerable challenge posed by high rates of bacterial drug resistance in the effective treatment of diarrheal RMs.
Collapse
Affiliation(s)
- Jie Wu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Quan Zhou
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Haoxuan Qi
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Weiqi Lan
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Shujian Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Shengzhi Yang
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China; Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China; Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.
| | - Anyun Zhang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610065, China.
| |
Collapse
|
33
|
Silva-Pilipich N, Beloki U, Salaberry L, Smerdou C. Self-Amplifying RNA: A Second Revolution of mRNA Vaccines against COVID-19. Vaccines (Basel) 2024; 12:318. [PMID: 38543952 PMCID: PMC10974399 DOI: 10.3390/vaccines12030318] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/12/2024] Open
Abstract
SARS-CoV-2 virus, the causative agent of COVID-19, has produced the largest pandemic in the 21st century, becoming a very serious health problem worldwide. To prevent COVID-19 disease and infection, a large number of vaccines have been developed and approved in record time, including new vaccines based on mRNA encapsulated in lipid nanoparticles. While mRNA-based vaccines have proven to be safe and effective, they are more expensive to produce compared to conventional vaccines. A special type of mRNA vaccine is based on self-amplifying RNA (saRNA) derived from the genome of RNA viruses, mainly alphaviruses. These saRNAs encode a viral replicase in addition to the antigen, usually the SARS-CoV-2 spike protein. The replicase can amplify the saRNA in transfected cells, potentially reducing the amount of RNA needed for vaccination and promoting interferon I responses that can enhance adaptive immunity. Preclinical studies with saRNA-based COVID-19 vaccines in diverse animal models have demonstrated the induction of robust protective immune responses, similar to conventional mRNA but at lower doses. Initial clinical trials have confirmed the safety and immunogenicity of saRNA-based vaccines in individuals that had previously received authorized COVID-19 vaccines. These findings have led to the recent approval of two of these vaccines by the national drug agencies of India and Japan, underscoring the promising potential of this technology.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Uxue Beloki
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Laura Salaberry
- Facultad de Ingeniería, Universidad ORT Uruguay, Montevideo 11100, Uruguay;
- Nanogrow Biotech, Montevideo 11500, Uruguay
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| |
Collapse
|
34
|
Belser JA, Kieran TJ, Mitchell ZA, Sun X, Mayfield K, Tumpey TM, Spengler JR, Maines TR. Key considerations to improve the normalization, interpretation and reproducibility of morbidity data in mammalian models of viral disease. Dis Model Mech 2024; 17:dmm050511. [PMID: 38440823 PMCID: PMC10941659 DOI: 10.1242/dmm.050511] [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: 09/15/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Viral pathogenesis and therapeutic screening studies that utilize small mammalian models rely on the accurate quantification and interpretation of morbidity measurements, such as weight and body temperature, which can vary depending on the model, agent and/or experimental design used. As a result, morbidity-related data are frequently normalized within and across screening studies to aid with their interpretation. However, such data normalization can be performed in a variety of ways, leading to differences in conclusions drawn and making comparisons between studies challenging. Here, we discuss variability in the normalization, interpretation, and presentation of morbidity measurements for four model species frequently used to study a diverse range of human viral pathogens - mice, hamsters, guinea pigs and ferrets. We also analyze findings aggregated from influenza A virus-infected ferrets to contextualize this discussion. We focus on serially collected weight and temperature data to illustrate how the conclusions drawn from this information can vary depending on how raw data are collected, normalized and measured. Taken together, this work supports continued efforts in understanding how normalization affects the interpretation of morbidity data and highlights best practices to improve the interpretation and utility of these findings for extrapolation to public health contexts.
Collapse
Affiliation(s)
- Jessica A. Belser
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Troy J. Kieran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Zoë A. Mitchell
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, USA
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Xiangjie Sun
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Kristin Mayfield
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Terrence M. Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jessica R. Spengler
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Taronna R. Maines
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| |
Collapse
|
35
|
Herron ICT, Laws TR, Nelson M. Marmosets as models of infectious diseases. Front Cell Infect Microbiol 2024; 14:1340017. [PMID: 38465237 PMCID: PMC10921895 DOI: 10.3389/fcimb.2024.1340017] [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: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Animal models of infectious disease often serve a crucial purpose in obtaining licensure of therapeutics and medical countermeasures, particularly in situations where human trials are not feasible, i.e., for those diseases that occur infrequently in the human population. The common marmoset (Callithrix jacchus), a Neotropical new-world (platyrrhines) non-human primate, has gained increasing attention as an animal model for a number of diseases given its small size, availability and evolutionary proximity to humans. This review aims to (i) discuss the pros and cons of the common marmoset as an animal model by providing a brief snapshot of how marmosets are currently utilized in biomedical research, (ii) summarize and evaluate relevant aspects of the marmoset immune system to the study of infectious diseases, (iii) provide a historical backdrop, outlining the significance of infectious diseases and the importance of developing reliable animal models to test novel therapeutics, and (iv) provide a summary of infectious diseases for which a marmoset model exists, followed by an in-depth discussion of the marmoset models of two studied bacterial infectious diseases (tularemia and melioidosis) and one viral infectious disease (viral hepatitis C).
Collapse
Affiliation(s)
- Ian C. T. Herron
- CBR Division, Defence Science and Technology Laboratory (Dstl), Salisbury, United Kingdom
| | | | | |
Collapse
|
36
|
Nazari-Vanani R, Negahdary M. Recent advances in electrochemical aptasensors and genosensors for the detection of pathogens. ENVIRONMENTAL RESEARCH 2024; 243:117850. [PMID: 38081349 DOI: 10.1016/j.envres.2023.117850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
In recent years, pathogenic microorganisms have caused significant mortality rates and antibiotic resistance and triggered exorbitant healthcare costs. These pathogens often have high transmission rates within human populations. Rapid diagnosis is crucial in controlling and reducing the spread of pathogenic infections. The diagnostic methods currently used against individuals infected with these pathogens include relying on outward symptoms, immunological-based and, some biomolecular ones, which mainly have limitations such as diagnostic errors, time-consuming processes, and high-cost platforms. Electrochemical aptasensors and genosensors have emerged as promising diagnostic tools for rapid, accurate, and cost-effective pathogen detection. These bio-electrochemical platforms have been optimized for diagnostic purposes by incorporating advanced materials (mainly nanomaterials), biomolecular technologies, and innovative designs. This review classifies electrochemical aptasensors and genosensors developed between 2021 and 2023 based on their use of different nanomaterials, such as gold-based, carbon-based, and others that employed other innovative assemblies without the use of nanomaterials. Inspecting the diagnostic features of various sensing platforms against pathogenic analytes can identify research gaps and open new avenues for exploration.
Collapse
Affiliation(s)
- Razieh Nazari-Vanani
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Negahdary
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, São Paulo, 05508-000, Brazil.
| |
Collapse
|
37
|
Kaplan BLF, Hoberman AM, Slikker W, Smith MA, Corsini E, Knudsen TB, Marty MS, Sobrian SK, Fitzpatrick SC, Ratner MH, Mendrick DL. Protecting Human and Animal Health: The Road from Animal Models to New Approach Methods. Pharmacol Rev 2024; 76:251-266. [PMID: 38351072 PMCID: PMC10877708 DOI: 10.1124/pharmrev.123.000967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/18/2023] [Accepted: 12/01/2023] [Indexed: 02/16/2024] Open
Abstract
Animals and animal models have been invaluable for our current understanding of human and animal biology, including physiology, pharmacology, biochemistry, and disease pathology. However, there are increasing concerns with continued use of animals in basic biomedical, pharmacological, and regulatory research to provide safety assessments for drugs and chemicals. There are concerns that animals do not provide sufficient information on toxicity and/or efficacy to protect the target population, so scientists are utilizing the principles of replacement, reduction, and refinement (the 3Rs) and increasing the development and application of new approach methods (NAMs). NAMs are any technology, methodology, approach, or assay used to understand the effects and mechanisms of drugs or chemicals, with specific focus on applying the 3Rs. Although progress has been made in several areas with NAMs, complete replacement of animal models with NAMs is not yet attainable. The road to NAMs requires additional development, increased use, and, for regulatory decision making, usually formal validation. Moreover, it is likely that replacement of animal models with NAMs will require multiple assays to ensure sufficient biologic coverage. The purpose of this manuscript is to provide a balanced view of the current state of the use of animal models and NAMs as approaches to development, safety, efficacy, and toxicity testing of drugs and chemicals. Animals do not provide all needed information nor do NAMs, but each can elucidate key pieces of the puzzle of human and animal biology and contribute to the goal of protecting human and animal health. SIGNIFICANCE STATEMENT: Data from traditional animal studies have predominantly been used to inform human health safety and efficacy. Although it is unlikely that all animal studies will be able to be replaced, with the continued advancement in new approach methods (NAMs), it is possible that sometime in the future, NAMs will likely be an important component by which the discovery, efficacy, and toxicity testing of drugs and chemicals is conducted and regulatory decisions are made.
Collapse
Affiliation(s)
- Barbara L F Kaplan
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Alan M Hoberman
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - William Slikker
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Mary Alice Smith
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Emanuela Corsini
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Thomas B Knudsen
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - M Sue Marty
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Sonya K Sobrian
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Suzanne C Fitzpatrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Marcia H Ratner
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Donna L Mendrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| |
Collapse
|
38
|
Liu G, Zhang M, Wu B, Zhang C, Wang Y, Han X, Wang R, Li L, Wei Y, Sun Y, Cao X, Wang Y, Li Y, Li M, Zhao G, Ke Y, Guo Z, Yin Q, Sun Y. A highly susceptible hACE2-transgenic mouse model for SARS-CoV-2 research. Front Microbiol 2024; 15:1348405. [PMID: 38389533 PMCID: PMC10883650 DOI: 10.3389/fmicb.2024.1348405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Several animal models have been used to assist the development of vaccines and therapeutics since the COVID-19 outbreak. Due to the lack of binding affinity of mouse angiotensin-converting enzyme II (ACE2) to the S protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), increasing the susceptibility of mice to SARS-CoV-2 infection was considered in several ways. Here, we generated a COVID-19 mouse model expressing human ACE2 (hACE2) under the control of the CAG promoter. Overexpression of hACE2 did not pose a significant effect on weight growth. After SARS-CoV-2 inoculation, mice showed obvious viral replication and production of inflammation within 7 days, with a gradual decrease in body weight until death. Virological testing found that the virus can replicate in the respiratory system, small intestine, and brain. Additionally, this mouse model was applied to compare two antibody drug candidates, the anti-RBD antibody (MW06) and the mouse CD24-conjugated anti-RBD antibody (mCD24-MW06). Differences in antiviral effects between these two antibodies can be demonstrated in this mouse model when a challenge dose that invalidates the anti-RBD antibody treatment was used. This study provided a new mouse model for studying SARS-CoV-2 pathogenesis and evaluating potential interventions.
Collapse
Affiliation(s)
- Gang Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Min Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Baolei Wu
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Cheng Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yan Wang
- SPF (Beijing) Biotechnology Co., Ltd., Baoding, China
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Rongjuan Wang
- Beijing Kohnoor Science & Technology Co. Ltd., Beijing, China
| | - Li Li
- SPF (Beijing) Biotechnology Co., Ltd., Baoding, China
| | - Yuwei Wei
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yali Sun
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Xiangwen Cao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yalan Li
- Beijing Kohnoor Science & Technology Co. Ltd., Beijing, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zhendong Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yansong Sun
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
39
|
Kwon T. Utilizing non-human primate models to combat recent COVID-19/SARS-CoV-2 and viral infectious disease outbreaks. J Med Primatol 2024; 53:e12689. [PMID: 38084001 DOI: 10.1111/jmp.12689] [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: 10/05/2023] [Revised: 11/01/2023] [Accepted: 12/01/2023] [Indexed: 02/13/2024]
Abstract
In recent times, global viral outbreaks and diseases, such as COVID-19 (SARS-CoV-2), Zika (ZIKV), monkeypox (MPOX), Ebola (EBOV), and Marburg (MARV), have been extensively documented. Swiftly deciphering the mechanisms underlying disease pathogenesis and devising vaccines or therapeutic interventions to curtail these outbreaks stand as paramount imperatives. Amidst these endeavors, animal models emerge as pivotal tools. Among these models, non-human primates (NHPs) hold a position of particular importance. Their proximity in evolutionary lineage and physiological resemblances to humans render them a primary model for comprehending human viral infections. This review encapsulates the pivotal role of various NHP species-such as rhesus macaques (Macaca mulatta), cynomolgus macaques (Macaca fascicularis), african green monkeys (Chlorocebus sabaeus/aethiops), pigtailed macaques (Macaca nemestrina/Macaca leonina), baboons (Papio hamadryas/Papio anubis), and common marmosets (Callithrix jacchus)-in investigations pertaining to the abovementioned viral outbreaks. These NHP models play a pivotal role in illuminating key aspects of disease dynamics, facilitating the development of effective countermeasures, and contributing significantly to our overall understanding of viral pathogenesis.
Collapse
Affiliation(s)
- Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si, Jeonbuk, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon, Korea
| |
Collapse
|
40
|
Kar M, Johnson KEE, Vanderheiden A, Elrod EJ, Floyd K, Geerling E, Stone ET, Salinas E, Banakis S, Wang W, Sathish S, Shrihari S, Davis-Gardner ME, Kohlmeier J, Pinto A, Klein R, Grakoui A, Ghedin E, Suthar MS. CD4+ and CD8+ T cells are required to prevent SARS-CoV-2 persistence in the nasal compartment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576505. [PMID: 38410446 PMCID: PMC10896337 DOI: 10.1101/2024.01.23.576505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
SARS-CoV-2 is the causative agent of COVID-19 and continues to pose a significant public health threat throughout the world. Following SARS-CoV-2 infection, virus-specific CD4+ and CD8+ T cells are rapidly generated to form effector and memory cells and persist in the blood for several months. However, the contribution of T cells in controlling SARS-CoV-2 infection within the respiratory tract are not well understood. Using C57BL/6 mice infected with a naturally occurring SARS-CoV-2 variant (B.1.351), we evaluated the role of T cells in the upper and lower respiratory tract. Following infection, SARS-CoV-2-specific CD4+ and CD8+ T cells are recruited to the respiratory tract and a vast proportion secrete the cytotoxic molecule Granzyme B. Using antibodies to deplete T cells prior to infection, we found that CD4+ and CD8+ T cells play distinct roles in the upper and lower respiratory tract. In the lungs, T cells play a minimal role in viral control with viral clearance occurring in the absence of both CD4+ and CD8+ T cells through 28 days post-infection. In the nasal compartment, depletion of both CD4+ and CD8+ T cells, but not individually, results in persistent and culturable virus replicating in the nasal compartment through 28 days post-infection. Using in situ hybridization, we found that SARS-CoV-2 infection persisted in the nasal epithelial layer of tandem CD4+ and CD8+ T cell-depleted mice. Sequence analysis of virus isolates from persistently infected mice revealed mutations spanning across the genome, including a deletion in ORF6. Overall, our findings highlight the importance of T cells in controlling virus replication within the respiratory tract during SARS-CoV-2 infection.
Collapse
|
41
|
Kuddus MA, Paul AK, Theparod T. Cost-effectiveness analysis of COVID-19 intervention policies using a mathematical model: an optimal control approach. Sci Rep 2024; 14:494. [PMID: 38177230 PMCID: PMC10766655 DOI: 10.1038/s41598-023-50799-6] [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: 08/30/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
COVID-19 is an infectious disease that causes millions of deaths worldwide, and it is the principal leading cause of morbidity and mortality in all nations. Although the governments of developed and developing countries are enforcing their universal control strategies, more precise and cost-effective single or combination interventions are required to control COVID-19 outbreaks. Using proper optimal control strategies with appropriate cost-effectiveness analysis is important to simulate, examine, and forecast the COVID-19 transmission phase. In this study, we developed a COVID-19 mathematical model and considered two important features including direct link between vaccination and latently population, and practical healthcare cost by separation of infections into Mild and Critical cases. We derived basic reproduction numbers and performed mesh and contour plots to explore the impact of different parameters on COVID-19 dynamics. Our model fitted and calibrated with number of cases of the COVID-19 data in Bangladesh as a case study to determine the optimal combinations of interventions for particular scenarios. We evaluated the cost-effectiveness of varying single and combinations of three intervention strategies, including transmission control, treatment, and vaccination, all within the optimal control framework of the single-intervention policies; enhanced transmission control is the most cost-effective and prompt in declining the COVID-19 cases in Bangladesh. Our finding recommends that a three-intervention strategy that integrates transmission control, treatment, and vaccination is the most cost-effective compared to single and double intervention techniques and potentially reduce the overall infections. Other policies can be implemented to control COVID-19 depending on the accessibility of funds and policymakers' judgments.
Collapse
Affiliation(s)
- Md Abdul Kuddus
- Department of Mathematics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Anip Kumar Paul
- Department of Mathematics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Thitiya Theparod
- Department of Mathematics, Mahasarakham University, Maha Sarakham, 44150, Thailand.
| |
Collapse
|
42
|
Scheithauer TPM, Montijn RC, Mieremet A. Gut microbe-host interactions in post-COVID syndrome: a debilitating or restorative partnership? Gut Microbes 2024; 16:2402544. [PMID: 39287023 PMCID: PMC11409505 DOI: 10.1080/19490976.2024.2402544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Post-COVID syndrome (PCS) patients have reported a wide range of symptoms, including fatigue, shortness of breath, and diarrhea. Particularly, the presence of gastrointestinal symptoms has led to the hypothesis that the gut microbiome is involved in the development and severity of PCS. The objective of this review is to provide an overview of the role of the gut microbiome in PCS by describing the microbial composition and microbial metabolites in COVID-19 and PCS. Moreover, host-microbe interactions via the microbiota-gut-brain (MGB) and the microbiota-gut-lung (MGL) axes are described. Furthermore, we explore the potential of therapeutically targeting the gut microbiome to support the recovery of PCS by reviewing preclinical model systems and clinical studies. Overall, current studies provide evidence that the gut microbiota is affected in PCS; however, diversity in symptoms and highly individual microbiota compositions suggest the need for personalized medicine. Gut-targeted therapies, including treatments with pre- and probiotics, have the potential to improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Torsten P M Scheithauer
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Roy C Montijn
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Arnout Mieremet
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
43
|
Hu W, Meng L, Wang C, Lu W, Tong X, Lin R, Xu T, Chen L, Cui A, Xu X, Li A, Tang J, Gao H, Pei Z, Zhang R, Wang Y, Wang Y, Han W, Jiang N, Xiong C, Feng Y, Lee K, Chen M. Spatiotemporal observations of host-pathogen interactions in mucosa during SARS-CoV-2 infection indicate a protective role of ILC2s. Microbiol Spectr 2023; 11:e0087823. [PMID: 37937994 PMCID: PMC10714800 DOI: 10.1128/spectrum.00878-23] [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: 05/15/2023] [Accepted: 09/30/2023] [Indexed: 11/09/2023] Open
Abstract
IMPORTANCE Our study revealed the spatial interaction between humanized ACE2 and pseudovirus expressing Spike, emphasizing the role of type 2 innate lymphoid cells during the initial phase of viral infection. These findings provide a foundation for the development of mucosal vaccines and other treatment approaches for both pre- and post-infection management of coronavirus disease 2019.
Collapse
Affiliation(s)
- Wei Hu
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Lu Meng
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chao Wang
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenhan Lu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Rui Lin
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Tao Xu
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - An Cui
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoqing Xu
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Anni Li
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia Tang
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongru Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenle Pei
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ruonan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wendong Han
- Biosafety Level 3 Laboratory, Shanghai Medical College Fudan University, Shanghai, China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Chenglong Xiong
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Kuinyu Lee
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mingquan Chen
- Department of Emergency Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Zumbrun EE, Zak SE, Lee ED, Bowling PA, Ruiz SI, Zeng X, Koehler JW, Delp KL, Bakken RR, Hentschel SS, Bloomfield HA, Ricks KM, Clements TL, Babka AM, Dye JM, Herbert AS. SARS-CoV-2 Aerosol and Intranasal Exposure Models in Ferrets. Viruses 2023; 15:2341. [PMID: 38140582 PMCID: PMC10747480 DOI: 10.3390/v15122341] [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: 11/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the worldwide COVID-19 pandemic. Animal models are extremely helpful for testing vaccines and therapeutics and for dissecting the viral and host factors that contribute to disease severity and transmissibility. Here, we report the assessment and comparison of intranasal and small particle (~3 µm) aerosol SARS-CoV-2 exposure in ferrets. The primary endpoints for analysis were clinical signs of disease, recovery of the virus in the upper respiratory tract, and the severity of damage within the respiratory tract. This work demonstrated that ferrets were productively infected with SARS-CoV-2 following either intranasal or small particle aerosol exposure. SARS-CoV-2 infection of ferrets resulted in an asymptomatic disease course following either intranasal or small particle aerosol exposure, with no clinical signs, significant weight loss, or fever. In both aerosol and intranasal ferret models, SARS-CoV-2 replication, viral genomes, and viral antigens were detected within the upper respiratory tract, with little to no viral material detected in the lungs. The ferrets exhibited a specific IgG immune response to the SARS-CoV-2 full spike protein. Mild pathological findings included inflammation, necrosis, and edema within nasal turbinates, which correlated to positive immunohistochemical staining for the SARS-CoV-2 virus. Environmental sampling was performed following intranasal exposure of ferrets, and SARS-CoV-2 genomic material was detected on the feeders and nesting areas from days 2-10 post-exposure. We conclude that both intranasal and small particle aerosol ferret models displayed measurable parameters that could be utilized for future studies, including transmission studies and testing SARS-CoV-2 vaccines and therapeutics.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Samantha E. Zak
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Eric D. Lee
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Philip A. Bowling
- Division of Veterinary Medicine, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Sara I. Ruiz
- Division of Bacteriology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Xiankun Zeng
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Jeffrey W. Koehler
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Korey L. Delp
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Russel R. Bakken
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Shannon S. Hentschel
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Holly A. Bloomfield
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Keersten M. Ricks
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Tamara L. Clements
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - April M. Babka
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - John M. Dye
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Andrew S. Herbert
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| |
Collapse
|
45
|
Condor Capcha JM, Kamiar A, Robleto E, Saad AG, Cui T, Wong A, Villano J, Zhong W, Pekosz A, Medina E, Cai R, Sha W, Ranek MJ, Webster KA, Schally AV, Jackson RM, Shehadeh LA. Growth hormone-releasing hormone receptor antagonist MIA-602 attenuates cardiopulmonary injury induced by BSL-2 rVSV-SARS-CoV-2 in hACE2 mice. Proc Natl Acad Sci U S A 2023; 120:e2308342120. [PMID: 37983492 PMCID: PMC10691341 DOI: 10.1073/pnas.2308342120] [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: 05/25/2023] [Accepted: 09/07/2023] [Indexed: 11/22/2023] Open
Abstract
COVID-19 pneumonia causes acute lung injury and acute respiratory distress syndrome (ALI/ARDS) characterized by early pulmonary endothelial and epithelial injuries with altered pulmonary diffusing capacity and obstructive or restrictive physiology. Growth hormone-releasing hormone receptor (GHRH-R) is expressed in the lung and heart. GHRH-R antagonist, MIA-602, has been reported to modulate immune responses to bleomycin lung injury and inflammation in granulomatous sarcoidosis. We hypothesized that MIA-602 would attenuate rVSV-SARS-CoV-2-induced pulmonary dysfunction and heart injury in a BSL-2 mouse model. Male and female K18-hACE2tg mice were inoculated with SARS-CoV-2/USA-WA1/2020, BSL-2-compliant recombinant VSV-eGFP-SARS-CoV-2-Spike (rVSV-SARS-CoV-2), or PBS, and lung viral load, weight loss, histopathology, and gene expression were compared. K18-hACE2tg mice infected with rVSV-SARS-CoV-2 were treated daily with subcutaneous MIA-602 or vehicle and conscious, unrestrained plethysmography performed on days 0, 3, and 5 (n = 7 to 8). Five days after infection mice were killed, and blood and tissues collected for histopathology and protein/gene expression. Both native SARS-CoV-2 and rVSV-SARS-CoV-2 presented similar patterns of weight loss, infectivity (~60%), and histopathologic changes. Daily treatment with MIA-602 conferred weight recovery, reduced lung perivascular inflammation/pneumonia, and decreased lung/heart ICAM-1 expression compared to vehicle. MIA-602 rescued altered respiratory rate, increased expiratory parameters (Te, PEF, EEP), and normalized airflow parameters (Penh and Rpef) compared to vehicle, consistent with decreased airway inflammation. RNASeq followed by protein analysis revealed heightened levels of inflammation and end-stage necroptosis markers, including ZBP1 and pMLKL induced by rVSV-SARS-CoV-2, that were normalized by MIA-602 treatment, consistent with an anti-inflammatory and pro-survival mechanism of action in this preclinical model of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Jose M. Condor Capcha
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
| | - Ali Kamiar
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
| | - Emely Robleto
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
| | - Ali G. Saad
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
| | - Tengjiao Cui
- Research Service, Miami Veterans Affairs Health System (VAHS), Miami, FL33125
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL33101
| | - Amanda Wong
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD21205
| | - Jason Villano
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD21205
| | - William Zhong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD21205
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD21205
| | - Edgar Medina
- Qualityminds Gesellschaft mit beschränkter Haftung (GmbH), Munchen, Munich81549, Germany
| | - Renzhi Cai
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Research Service, Miami Veterans Affairs Health System (VAHS), Miami, FL33125
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL33101
| | - Wei Sha
- Research Service, Miami Veterans Affairs Health System (VAHS), Miami, FL33125
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL33101
| | - Mark J. Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD21205
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD21205
| | - Keith A. Webster
- Integene International Holdings, Miami, FL33179
- Baylor College of Medicine, Houston, TX77030
| | - Andrew V. Schally
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Research Service, Miami Veterans Affairs Health System (VAHS), Miami, FL33125
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL33101
| | - Robert M. Jackson
- Research Service, Miami Veterans Affairs Health System (VAHS), Miami, FL33125
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL33101
| | - Lina A. Shehadeh
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL33136
| |
Collapse
|
46
|
Jack KM, Kulick NK. Primate field research during a pandemic: Lessons learned from the SARS-CoV-2 outbreak. Am J Primatol 2023; 85:e23551. [PMID: 37706674 DOI: 10.1002/ajp.23551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023]
Abstract
The COVID-19 pandemic abruptly halted most primate field research in early 2020. While international travel bans and regional travel restrictions made continuing primate field research impossible early on in the pandemic, ethical concerns of transmitting the virus from researchers to primates and surrounding human communities informed decisions regarding the timing of resuming research. Between June and September 2020, we surveyed field primatologists regarding the impacts of the pandemic on their research. We received 90 completed surveys from respondents residing in 21 countries, though most were from the United States and Canada. These data provide a valuable window into the perspectives and actions taken by researchers during the early stages of the pandemic as events were still unfolding. Only 2.4% of projects reported continuing research as usual, 33.7% continued with some decrease in productivity, 42.2% reported postponing research projects, and 21.7% reported canceling projects or postponing research indefinitely. Respondents most severely impacted by the pandemic were those establishing new field sites and graduate students whose projects were postponed or canceled due to pandemic-related shutdowns. Fears about increased poaching, the inability to pay local assistants, frozen research funds, declining habituation, disruptions to data collection, and delays in student projects were among the top concerns of respondents. Nearly all the projects able to continue research in any capacity during the early months of the pandemic were run by or employed primate habitat country primatologists. This finding is a major lesson learned from the pandemic; without habitat country scientists, primate research is not sustainable.
Collapse
Affiliation(s)
- Katharine M Jack
- Department of Anthropology, Tulane University, New Orleans, Louisiana, USA
| | - Nelle K Kulick
- Department of Anthropology, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
47
|
Brandenburg K, Ferrer-Espada R, Martinez-de-Tejada G, Nehls C, Fukuoka S, Mauss K, Weindl G, Garidel P. A Comparison between SARS-CoV-2 and Gram-Negative Bacteria-Induced Hyperinflammation and Sepsis. Int J Mol Sci 2023; 24:15169. [PMID: 37894850 PMCID: PMC10607443 DOI: 10.3390/ijms242015169] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis is a life-threatening condition caused by the body's overwhelming response to an infection, such as pneumonia or urinary tract infection. It occurs when the immune system releases cytokines into the bloodstream, triggering widespread inflammation. If not treated, it can lead to organ failure and death. Unfortunately, sepsis has a high mortality rate, with studies reporting rates ranging from 20% to over 50%, depending on the severity and promptness of treatment. According to the World Health Organization (WHO), the annual death toll in the world is about 11 million. One of the main toxins responsible for inflammation induction are lipopolysaccharides (LPS, endotoxin) from Gram-negative bacteria, which rank among the most potent immunostimulants found in nature. Antibiotics are consistently prescribed as a part of anti-sepsis-therapy. However, antibiotic therapy (i) is increasingly ineffective due to resistance development and (ii) most antibiotics are unable to bind and neutralize LPS, a prerequisite to inhibit the interaction of endotoxin with its cellular receptor complex, namely Toll-like receptor 4 (TLR4)/MD-2, responsible for the intracellular cascade leading to pro-inflammatory cytokine secretion. The pandemic virus SARS-CoV-2 has infected hundreds of millions of humans worldwide since its emergence in 2019. The COVID-19 (Coronavirus disease-19) caused by this virus is associated with high lethality, particularly for elderly and immunocompromised people. As of August 2023, nearly 7 million deaths were reported worldwide due to this disease. According to some reported studies, upregulation of TLR4 and the subsequent inflammatory signaling detected in COVID-19 patients "mimics bacterial sepsis". Furthermore, the immune response to SARS-CoV-2 was described by others as "mirror image of sepsis". Similarly, the cytokine profile in sera from severe COVID-19 patients was very similar to those suffering from the acute respiratory distress syndrome (ARDS) and sepsis. Finally, the severe COVID-19 infection is frequently accompanied by bacterial co-infections, as well as by the presence of significant LPS concentrations. In the present review, we will analyze similarities and differences between COVID-19 and sepsis at the pathophysiological, epidemiological, and molecular levels.
Collapse
Affiliation(s)
- Klaus Brandenburg
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
| | - Raquel Ferrer-Espada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
| | - Christian Nehls
- Forschungszentrum Borstel, FG Biophysik, Parkallee 10, 23845 Borstel, Germany;
| | - Satoshi Fukuoka
- National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu 761-0395, Japan;
| | - Karl Mauss
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
- Sylter Klinik Karl Mauss, Dr.-Nicolas-Strasse 3, 25980 Westerland (Sylt), Germany
| | - Günther Weindl
- Pharmazeutisches Institut, Abteilung Pharmakologie und Toxikologie, Universität Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany;
| | - Patrick Garidel
- Physikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, 06108 Halle (Saale), Germany
| |
Collapse
|
48
|
Hoffmann M, Wong LYR, Arora P, Zhang L, Rocha C, Odle A, Nehlmeier I, Kempf A, Richter A, Halwe NJ, Schön J, Ulrich L, Hoffmann D, Beer M, Drosten C, Perlman S, Pöhlmann S. Omicron subvariant BA.5 efficiently infects lung cells. Nat Commun 2023; 14:3500. [PMID: 37311762 PMCID: PMC10262933 DOI: 10.1038/s41467-023-39147-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
The SARS-CoV-2 Omicron subvariants BA.1 and BA.2 exhibit reduced lung cell infection relative to previously circulating SARS-CoV-2 variants, which may account for their reduced pathogenicity. However, it is unclear whether lung cell infection by BA.5, which displaced these variants, remains attenuated. Here, we show that the spike (S) protein of BA.5 exhibits increased cleavage at the S1/S2 site and drives cell-cell fusion and lung cell entry with higher efficiency than its counterparts from BA.1 and BA.2. Increased lung cell entry depends on mutation H69Δ/V70Δ and is associated with efficient replication of BA.5 in cultured lung cells. Further, BA.5 replicates in the lungs of female Balb/c mice and the nasal cavity of female ferrets with much higher efficiency than BA.1. These results suggest that BA.5 has acquired the ability to efficiently infect lung cells, a prerequisite for causing severe disease, suggesting that evolution of Omicron subvariants can result in partial loss of attenuation.
Collapse
Affiliation(s)
- Markus Hoffmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany.
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany.
| | - Lok-Yin Roy Wong
- Departments of Microbiology and Immunology, BSB 3-712, University of Iowa, Iowa City, IA, USA
| | - Prerna Arora
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Lu Zhang
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Cheila Rocha
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Abby Odle
- Departments of Microbiology and Immunology, BSB 3-712, University of Iowa, Iowa City, IA, USA
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Anja Richter
- Institute of Virology, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Nico Joel Halwe
- Institut für Virusdiagnostik (IVD), Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Jacob Schön
- Institut für Virusdiagnostik (IVD), Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Lorenz Ulrich
- Institut für Virusdiagnostik (IVD), Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Donata Hoffmann
- Institut für Virusdiagnostik (IVD), Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Martin Beer
- Institut für Virusdiagnostik (IVD), Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Stanley Perlman
- Departments of Microbiology and Immunology, BSB 3-712, University of Iowa, Iowa City, IA, USA
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany.
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany.
| |
Collapse
|
49
|
Yang B, Liu C, Ju X, Wu B, Wang Z, Dong F, Yu Y, Hou X, Fang M, Gao F, Guo X, Gui Y, Ding Q, Li W. A tissue specific-infection mouse model of SARS-CoV-2. Cell Discov 2023; 9:43. [PMID: 37080957 PMCID: PMC10117269 DOI: 10.1038/s41421-023-00536-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/06/2023] [Indexed: 04/22/2023] Open
Abstract
Animal models play crucial roles in the rapid development of vaccines/drugs for the prevention and therapy of COVID-19, but current models have some deficits when studying the pathogenesis of SARS-CoV-2 on some special tissues or organs. Here, we generated a human ACE2 and SARS-CoV-2 NF/F knockin mouse line that constitutively expresses human ACE2 and specifically expresses SARS-CoV-2 N gene induced by Cre-recombinase. By crossing with Cre transgenic lines allowing for lung-specific and constitutive expression, we generated lung-specific (Sftpc-hACE2-NF/F) and constitutive SARS-CoV-2 N (EIIa-hACE2-NF/F) expressing mice. Upon intranasal infection with a SARS-CoV-2 GFP/ΔN strain which can only replicate in SARS-CoV-2 N expressed cells, we demonstrated that both the Sftpc-hACE2-NF/F and EIIa-hACE2-NF/F mice support viral replication. Consistent with our design, viral replication was limited to the lung tissues in Sftpc-hACE2-NF/F mice, while the EIIa-hACE2-NF/F mice developed infections in multiple tissues. Furthermore, our model supports different SARS-CoV-2 variants infection, and it can be successfully used to evaluate the effects of therapeutic monoclonal antibodies (Ab1F11) and antiviral drugs (Molnupiravir). Finally, to test the effect of SARS-CoV-2 infection on male reproduction, we generated Sertoli cell-specific SARS-CoV-2 N expressed mice by crossing with AMH-Cre transgenic line. We found that SARS-CoV-2 GFP/ΔN strain could infect Sertoli cells, led to spermatogenic defects due to the destruction of blood-testis barrier. Overall, combining with different tissue-specific Cre transgenic lines, the human ACE2 and SARS-CoV-2 NF/F line enables us to evaluate antivirals in vivo and study the pathogenesis of SARS-CoV-2 on some special tissues or organs.
Collapse
Affiliation(s)
- Bo Yang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chao Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaohui Ju
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuangfei Wang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Fucheng Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanying Yu
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohui Hou
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong, China.
| | - Qiang Ding
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China.
| | - Wei Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
50
|
Szpiro L, Bourgeay C, Hoareau AL, Julien T, Menard C, Marie Y, Rosa-Calatrava M, Moules V. Antiviral Activity of Active Materials: Standard and Finger-Pad-Based Innovative Experimental Approaches. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2889. [PMID: 37049183 PMCID: PMC10096329 DOI: 10.3390/ma16072889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 06/19/2023]
Abstract
Environmental surfaces, including high-touch surfaces (HITS), bear a high risk of becoming fomites and can participate in viral dissemination through contact and transmission to other persons, due to the capacity of viruses to persist on such contaminated surface before being transferred to hands or other supports at sufficient concentration to initiate infection through direct contact. Interest in the development of self-decontaminating materials as additional safety measures towards preventing viral infectious disease transmission has been growing. Active materials are expected to reduce the viral charge on surfaces over time and consequently limit viral transmission capacity through direct contact. In this study, we compared antiviral activities obtained using three different experimental procedures by assessing the survival of an enveloped virus (influenza virus) and non-enveloped virus (feline calicivirus) over time on a reference surface and three active materials. Our data show that experimental test conditions can have a substantial impact of over 1 log10 on the antiviral activity of active material for the same contact period, depending on the nature of the virus. We then developed an innovative and reproducible approach based on finger-pad transfer to evaluate the antiviral activity of HITS against a murine norovirus inoculum under conditions closely reflecting real-life surface exposure.
Collapse
Affiliation(s)
- Lea Szpiro
- VirHealth SAS, Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
| | - Clara Bourgeay
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Associated Laboratory RespiVir, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Associated Laboratory RespiVir, University Laval, Québec, QC G1V 0A6, Canada
| | - Alexandre Loic Hoareau
- VirHealth SAS, Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
| | - Thomas Julien
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Associated Laboratory RespiVir, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Associated Laboratory RespiVir, University Laval, Québec, QC G1V 0A6, Canada
| | - Camille Menard
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Associated Laboratory RespiVir, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Associated Laboratory RespiVir, University Laval, Québec, QC G1V 0A6, Canada
| | - Yana Marie
- VirHealth SAS, Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Associated Laboratory RespiVir, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Associated Laboratory RespiVir, University Laval, Québec, QC G1V 0A6, Canada
| | - Vincent Moules
- VirHealth SAS, Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
- Joint Technology Research Laboratory on Pathogenic Respiratory Viruses (PRV TEchLab), Innovation Centre Lyonbiopole, 321 Avenue Jean Jaurès, 69007 Lyon, France
| |
Collapse
|