1
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [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: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
2
|
Sefik E, Xiao T, Chiorazzi M, Odell I, Zhang F, Agrawal K, Micevic G, Flavell RA. Engineering Mice to Study Human Immunity. Annu Rev Immunol 2025; 43:451-487. [PMID: 40020225 DOI: 10.1146/annurev-immunol-082523-124415] [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] [Indexed: 04/26/2025]
Abstract
Humanized mice, which carry a human hematopoietic and immune system, have greatly advanced our understanding of human immune responses and immunological diseases. These mice are created via the transplantation of human hematopoietic stem and progenitor cells into immunocompromised murine hosts further engineered to support human hematopoiesis and immune cell growth. This article explores genetic modifications in mice that enhance xeno-tolerance, promote human hematopoiesis and immunity, and enable xenotransplantation of human tissues with resident immune cells. We also discuss genetic editing of the human immune system, provide examples of how humanized mice with humanized organs model diseases for mechanistic studies, and highlight the roles of these models in advancing knowledge of organ biology, immune responses to pathogens, and preclinical drugs tested for cancer treatment. The integration of multi-omics and state-of-the art approaches with humanized mouse models is crucial for bridging existing human data with causality and promises to significantly advance mechanistic studies.
Collapse
Affiliation(s)
- Esen Sefik
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
| | - Tianli Xiao
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Chiorazzi
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ian Odell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Fengrui Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kriti Agrawal
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Computational Biology and Bioinformatics Program, Yale University, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Goran Micevic
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Detrille A, Huvelle S, van Gils MJ, Geara T, Pascal Q, Snitselaar J, Bossevot L, Cavarelli M, Dereuddre-Bosquet N, Relouzat F, Contreras V, Chapon C, Caillé F, Sanders RW, Le Grand R, Naninck T. Whole-body visualization of SARS-CoV-2 biodistribution in vivo by immunoPET imaging in non-human primates. Nat Commun 2025; 16:2816. [PMID: 40118860 PMCID: PMC11928647 DOI: 10.1038/s41467-025-58173-y] [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: 10/10/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
The COVID-19 pandemic has caused at least 780 million cases globally. While available treatments and vaccines have reduced the mortality rate, spread and evolution of the virus are ongoing processes. Despite extensive research, the long-term impact of SARS-CoV-2 infection is still poorly understood and requires further investigation. Routine analysis provides limited access to the tissues of patients, necessitating alternative approaches to investigate viral dissemination in the organism. We address this issue by implementing a whole-body in vivo imaging strategy to longitudinally assess the biodistribution of SARS-CoV-2. We demonstrate in a COVID-19 non-human primate model that a single injection of radiolabeled [89Zr]COVA1-27-DFO human monoclonal antibody targeting a preserved epitope of the SARS-CoV-2 spike protein allows longitudinal tracking of the virus by positron emission tomography with computed tomography (PET/CT). Convalescent animals exhibit a persistent [89Zr]COVA1-27-DFO PET signal in the lungs, as well as in the brain, three months following infection. This imaging approach also allows viral detection in various organs, including the airways and kidneys, of exposed animals during the acute infection phase. Overall, the technology we developed offers a comprehensive assessment of SARS-CoV-2 distribution in vivo and provides a promising approach for the non-invasive study of long-COVID pathophysiology.
Collapse
Affiliation(s)
- Alexandra Detrille
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Steve Huvelle
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Orsay, France
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Tatiana Geara
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Quentin Pascal
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Jonne Snitselaar
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Catherine Chapon
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Fabien Caillé
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Orsay, France
| | - Rogier W Sanders
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France.
| |
Collapse
|
4
|
Breugem TI, Riesebosch S, Zhang J, Mykytyn AZ, Krabbendam L, Groen N, Baptista Varela S, Schipper D, van den Doel PB, van Acker R, Stadhouders R, Lamers MM, Haagmans BL. Variable DPP4 expression in multiciliated cells of the human nasal epithelium as a determinant for MERS-CoV tropism. Proc Natl Acad Sci U S A 2025; 122:e2410630122. [PMID: 40048293 PMCID: PMC11929475 DOI: 10.1073/pnas.2410630122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/21/2024] [Indexed: 03/25/2025] Open
Abstract
Transmissibility of respiratory viruses is a complex viral trait that is intricately linked to tropism. Several highly transmissible viruses, including severe acute respiratory syndrome coronavirus 2 and Influenza viruses, specifically target multiciliated cells in the upper respiratory tract to facilitate efficient human-to-human transmission. In contrast, the zoonotic Middle East respiratory syndrome coronavirus (MERS-CoV) generally transmits poorly between humans, which is largely attributed to the absence of its receptor dipeptidyl peptidase 4 (DPP4) in the upper respiratory tract. At the same time, MERS-CoV epidemiology is characterized by occasional superspreading events, suggesting that some individuals can disseminate this virus effectively. Here, we utilized well-differentiated human pulmonary and nasal airway organoid-derived cultures to further delineate the respiratory tropism of MERS-CoV. We find that MERS-CoV replicated to high titers in both pulmonary and nasal airway cultures. Using single-cell messenger-RNA sequencing, immunofluorescence, and immunohistochemistry, we show that MERS-CoV preferentially targeted multiciliated cells, leading to loss of ciliary coverage. MERS-CoV cellular tropism was dependent on the differentiation of the organoid-derived cultures, and replication efficiency varied considerably between donors. Similarly, variable and focal expression of DPP4 was revealed in human nose tissues. This study indicates that the upper respiratory tract tropism of MERS-CoV may vary between individuals due to differences in DPP4 expression, providing an explanation for the unpredictable transmission pattern of MERS-CoV.
Collapse
Affiliation(s)
- Tim I. Breugem
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Samra Riesebosch
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Jingshu Zhang
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Anna Z. Mykytyn
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Lisette Krabbendam
- Pulmonary Medicine Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Nathalie Groen
- Single Cell Discoveries, Utrecht3584 BW, The Netherlands
| | - Sivana Baptista Varela
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Debby Schipper
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Petra B. van den Doel
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Romy van Acker
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Ralph Stadhouders
- Pulmonary Medicine Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| | - Mart M. Lamers
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Bart L. Haagmans
- Viroscience Department, Erasmus University Medical Center, Rotterdam3015 GD, The Netherlands
| |
Collapse
|
5
|
Dogadov DI, Kyuregyan KK, Minosyan AA, Goncharenko AM, Shmat EV, Mikhailov MI. [Acute respiratory viral infections in monkeys]. Vopr Virusol 2025; 70:7-24. [PMID: 40233333 DOI: 10.36233/0507-4088-293] [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: 02/03/2025] [Indexed: 04/17/2025]
Abstract
Acute respiratory viral infections (ARVI) are one of the most significant infections affecting the breeding of monkeys, especially among imported and captive primates. Respiratory diseases are also an important cause of morbidity and mortality in wild populations, and most of these infections can affect humans. Many anthropoid species, including apes, are susceptible to ARVI. Outbreaks of spontaneous respiratory infections have been described in many zoos and primatological centers around the world. Moreover, the study of spontaneous and experimental infection in laboratory primates provides an invaluable source of information on the biology and pathogenesis of ARVI and remains an indispensable tool for testing vaccines and drugs. The aim of this literature review was to summarize and analyze published data on the circulation of ARVI causative agents (parainfluenza viruses, adenoviruses, respiratory syncytial virus, influenza viruses, rhinoviruses, coronaviruses, metapneumoviruses, bocaviruses) among wild and captive primates, as well as the results of experimental modeling these infections in monkeys.
Collapse
Affiliation(s)
- D I Dogadov
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - K K Kyuregyan
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia
| | - A A Minosyan
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - A M Goncharenko
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - E V Shmat
- Sochi Institute (branch) of the Federal State Autonomous Educational Institution of Higher Education Peoples' Friendship University of Russia named after Patrice Lumumba
| | - M I Mikhailov
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| |
Collapse
|
6
|
Ma ZM, Olstad KJ, Van Rompay KKA, Iyer SS, Miller CJ, Reader JR. Pulmonary lymphoid tissue induced after SARS-CoV-2 infection in rhesus macaques. Front Immunol 2025; 16:1533050. [PMID: 40145084 PMCID: PMC11937022 DOI: 10.3389/fimmu.2025.1533050] [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: 11/22/2024] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Lung diseases are widespread worldwide. Pulmonary immunity plays a vital role against lung pathogens, including SARS-CoV-2 infection. Understanding the pathogenesis, including the development of local immune responses to infection, is fundamental for developing interventions to control the viral infection. Methods Using immunohistochemistry, we investigated the distribution of immune cells in the lungs of rhesus macaques experimentally infected with SARS-CoV-2 and euthanized 11-14 days later. Results Tertiary lymphoid tissue was found in all SARS-CoV-2 infected animals. The number (13.9 vs 1.5 iPLT number/ lung cm2), size (25992 vs 13946 µm2) and total area (0.46 vs 0.02 mm2 iPLT/ lung cm2) of the lymphoid tissue aggregations were significantly higher in SARS-CoV-2 infected animals than that of normal controls. This induced pulmonary lymphoid tissues comprised B cells, T cells, CD169 macrophages, and follicular dendritic cells with evidence of lymphocyte priming and differentiation. Discussion The results suggest local immunity plays an important role in the SARS-CoV-2 infection. Further study of pulmonary immunity could lead to new interventions to develop vaccine strategies and discover new immune-regulatory biomarkers in monitoring and controlling SARS-CoV-2 infection and other lung diseases.
Collapse
Affiliation(s)
- Zhong-Min Ma
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
| | - Katherine J. Olstad
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California (UC) Davis, Davis, CA, United States
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California (UC) Davis, Davis, CA, United States
| | - Smita S. Iyer
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California (UC) Davis, Davis, CA, United States
- Center for Immunology and Infectious Diseases, University of California (UC) Davis, Davis, CA, United States
| | - Christopher J. Miller
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California (UC) Davis, Davis, CA, United States
- Center for Immunology and Infectious Diseases, University of California (UC) Davis, Davis, CA, United States
| | - J. Rachel Reader
- California National Primate Research Center, University of California (UC) Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California (UC) Davis, Davis, CA, United States
| |
Collapse
|
7
|
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
|
8
|
Yang Q, Kelkar A, Manicassamy B, Neelamegham S. Conserved role of spike S2 domain N-glycosylation across betacoronaviruses. NPJ VIRUSES 2025; 3:4. [PMID: 40295734 PMCID: PMC11762317 DOI: 10.1038/s44298-024-00085-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/10/2024] [Indexed: 04/30/2025]
Abstract
Besides acting as an immunological shield, the N-glycans of SARS-CoV-2 are also critical for viral life cycle. As the S2 subunit of spike is highly conserved across betacoronaviruses, we determined the functional significance of the five 'stem N-glycans' located in S2 between N1098-N1194. Studies were performed with 31 Asn-to-Gln mutants, betacoronavirus virus-like particles and single-cycle viral replicons. Deletions of stem N-glycans enhanced S1 shedding from trimeric spike, reduced ACE2 binding and abolished syncytia formation. When three or more N-glycans were deleted, spike expression on cell surface and incorporation into virions was both reduced. Viral entry function was progressively lost upon deleting the N1098 glycan in combination with additional glycosite modifications. In addition to SARS-CoV-2, deleting stem N-glycans in SARS-CoV and MERS-CoV spike also prevented viral entry into target cells. These data suggest multiple functional roles for the stem N-glycans, and evolutionarily conserved properties for these complex carbohydrates across human betacoronaviruses.
Collapse
Affiliation(s)
- Qi Yang
- Chemical & Biological Engineering, State University of New York, Buffalo, NY, USA
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY, USA
| | - Anju Kelkar
- Chemical & Biological Engineering, State University of New York, Buffalo, NY, USA
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY, USA
| | | | - Sriram Neelamegham
- Chemical & Biological Engineering, State University of New York, Buffalo, NY, USA.
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY, USA.
- Biomedical Engineering, State University of New York, Buffalo, NY, USA.
- Medicine, State University of New York, Buffalo, NY, USA.
- Clinical & Translational Research Center, Buffalo, NY, USA.
| |
Collapse
|
9
|
Beltrami VA, Martins FRB, Martins DG, Queiroz-Junior CM, Félix FB, Resende LC, Santos FRDS, Lacerda LDSB, Costa VRDM, da Silva WN, Guimaraes PPG, Guimaraes G, Soriani FM, Teixeira MM, Costa VV, Pinho V. Selective phosphodiesterase 4 inhibitor roflumilast reduces inflammation and lung injury in models of betacoronavirus infection in mice. Inflamm Res 2025; 74:24. [PMID: 39862252 DOI: 10.1007/s00011-024-01985-3] [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: 07/31/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE We aimed to understand the potential therapeutic and anti-inflammatory effects of the phosphodiesterase-4 (PDE4) inhibitor roflumilast in models of pulmonary infection caused by betacoronaviruses. METHODS Mice were infected intranasally with murine hepatitis virus (MHV-3) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Roflumilast was given to MHV-3-infected mice therapeutically at doses of 1 mg/kg or 10 mg/kg, or prophylactically at 10 mg/kg. In SARS-CoV-2-infected mice, roflumilast was given therapeutically at a dose of 10 mg/kg. Lung histopathology, chemokines (CXCL-1 and CCL2), cytokines (IL-1β, IL-6, TNF, IFN-γ, IL-10 and TGFβ), neutrophil immunohistochemical staining (Ly6G+ cells), macrophage immunofluorescence staining (F4/80+ cells), viral titration plaque assay, real-time PCR virus detection, and blood cell counts were examined. RESULTS Therapeutic treatment with roflumilast at 10 mg/kg reduced lung injury in SARS-CoV-2 or MHV-3-infected mice without compromising viral clearance. In MHV-3-infected mice, reduced lung injury was associated with decreased chemokines levels, prevention of neutrophil aggregates and reduced macrophage accumulation in the lung tissue. However, the prophylactic treatment strategy with roflumilast increased lung injury in MHV-3-infected mice. CONCLUSION Our findings indicate that therapeutic treatment with roflumilast reduced lung injury in MHV-3 and SARS-CoV-2 lung infections. Given the protection induced by roflumilast in inflammation, PDE4 targeting could be a promising therapeutic avenue worth exploring following severe viral infections of the lung.
Collapse
Affiliation(s)
- Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Flávia Rayssa Braga Martins
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Franciel Batista Félix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Letícia Cassiano Resende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Larisse de Souza Barbosa Lacerda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Victor Rodrigues de Melo Costa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Walison Nunes da Silva
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Pedro Pires Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Frederico Marianetti Soriani
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
10
|
Xiao F, Hu J, Xu M, Wang D, Shen X, Zhang H, Miao J, Cai H, Wang J, Liu Y, Xiao S, Zhu L. Animal Models for Human-Pathogenic Coronavirus and Animal Coronavirus Research. Viruses 2025; 17:100. [PMID: 39861889 PMCID: PMC11768759 DOI: 10.3390/v17010100] [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: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Coronavirus epidemics have posed a serious threat to both human and animal health. To combat emerging infectious diseases caused by coronaviruses, various animal infection models have been developed and applied in research, including non-human primate models, ferret models, hamster models, mouse models, and others. Moreover, new approaches have been utilized to develop animal models that are more susceptible to infection. These approaches include using viral delivery methods to induce the expression of viral receptors in mouse tissues and employing gene-editing techniques to create genetically modified mice. This has led to the successful establishment of infection models for multiple coronaviruses, significantly advancing related research. In contrast, livestock and pets that can be infected by animal coronaviruses provide valuable insights when used as infection models, enabling the collection of accurate clinical data through the analysis of post-infection pathological features. However, despite the potential insights, there is a paucity of research data pertaining to these infection models. In this review, we provide a detailed overview of recent progress in the development of animal models for coronaviruses that cause diseases in both humans and animals and suggest ways in which animal models can be adapted to further enhance their value in research.
Collapse
Affiliation(s)
- Fenglian Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Traditional Chinese Medicine and Health School, Nanfang College, Guangzhou 510970, China
| | - Jincheng Hu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Minsheng Xu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Di Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Xiaoyan Shen
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Hua Zhang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jie Miao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Haodong Cai
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jihui Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Yaqing Liu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Shan Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Longchao Zhu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
11
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [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: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
12
|
Horton K, Wing PAC, Jackson CL, McCormick CJ, Carroll MP, Lucas JS. Interplay between respiratory viruses and cilia in the airways. Eur Respir Rev 2025; 34:240224. [PMID: 40107662 PMCID: PMC11920889 DOI: 10.1183/16000617.0224-2024] [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: 10/03/2024] [Accepted: 01/19/2025] [Indexed: 03/22/2025] Open
Abstract
The airway epithelium is the first point of contact for inhaled pathogens. The role of epithelial cells in clearance, infection and colonisation of bacteria is established. The interactions of respiratory viruses and cilia is less understood, but viruses are known to target ciliated epithelial cells for entry, replication and dissemination. Furthermore, some respiratory viruses impair and/or enhance ciliary activity. This review examines what is known about the interactions between cilia and viral infection and how respiratory viruses effect cilia function with subsequent consequences for human health. We discuss the models which can be used to investigate the relationship between respiratory viruses and the host airway.
Collapse
Affiliation(s)
- Katie Horton
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- These authors contributed equally to this work
| | - Peter A C Wing
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- These authors contributed equally to this work
| | - Claire L Jackson
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- These authors contributed equally to this work
| | - Christopher J McCormick
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Mary P Carroll
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
13
|
Matsumoto Y, Honda T, Yasui F, Endo A, Sanada T, Toyama S, Takagi A, Munakata T, Kono R, Yamaji K, Yamamoto N, Saeki Y, Kohara M. Generation of a SARS-CoV-2-susceptible mouse model using adenovirus vector expressing human angiotensin-converting enzyme 2 driven by an elongation factor 1α promoter with leftward orientation. Front Immunol 2024; 15:1440314. [PMID: 39717778 PMCID: PMC11663739 DOI: 10.3389/fimmu.2024.1440314] [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: 05/29/2024] [Accepted: 11/08/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction To analyze the molecular pathogenesis of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), a small animal model such as mice is needed: human angiotensin converting enzyme 2 (hACE2), the receptor of SARS-CoV-2, needs to be expressed in the respiratory tract of mice. Methods We conferred SARS-CoV-2 susceptibility in mice by using an adenoviral vector expressing hACE2 driven by an elongation factor 1α (EF1α) promoter with a leftward orientation. Results In this model, severe pneumonia like human COVID-19 was observed in SARS-CoV-2-infected mice, which was confirmed by dramatic infiltration of inflammatory cells in the lung with efficient viral replication. An early circulating strain of SARS-CoV-2 caused the most severe weight loss when compared to SARS-CoV-2 variants such as Alpha, Beta and Gamma, although histopathological findings, viral replication, and cytokine expression characteristics were comparable. Discussion We found that a distinct proteome of an early circulating strain infected lung characterized by elevated complement activation and blood coagulation, which were mild in other variants, can contribute to disease severity. Unraveling the specificity of early circulating SARS-CoV-2 strains is important in elucidating the origin of the pandemic.
Collapse
Affiliation(s)
- Yusuke Matsumoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Tomoko Honda
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Fumihiko Yasui
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akinori Endo
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takahiro Sanada
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sakiko Toyama
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Asako Takagi
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tsubasa Munakata
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Risa Kono
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenzaburo Yamaji
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Naoki Yamamoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasushi Saeki
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
14
|
Kirk NM, Liang Y, Ly H. Pathogenesis and virulence of coronavirus disease: Comparative pathology of animal models for COVID-19. Virulence 2024; 15:2316438. [PMID: 38362881 PMCID: PMC10878030 DOI: 10.1080/21505594.2024.2316438] [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: 10/20/2023] [Accepted: 02/04/2024] [Indexed: 02/17/2024] Open
Abstract
Animal models that can replicate clinical and pathologic features of severe human coronavirus infections have been instrumental in the development of novel vaccines and therapeutics. The goal of this review is to summarize our current understanding of the pathogenesis of coronavirus disease 2019 (COVID-19) and the pathologic features that can be observed in several currently available animal models. Knowledge gained from studying these animal models of SARS-CoV-2 infection can help inform appropriate model selection for disease modelling as well as for vaccine and therapeutic developments.
Collapse
Affiliation(s)
- Natalie M. Kirk
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| |
Collapse
|
15
|
Nisar KS, Naz I, Raja MAZ, Shoaib M. Intelligent computing framework to analyze the transmission risk of COVID-19: Meyer wavelet artificial neural networks. Comput Biol Chem 2024; 113:108234. [PMID: 39395247 DOI: 10.1016/j.compbiolchem.2024.108234] [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: 02/28/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024]
Abstract
The optimum control methods for the epidemiology of the COVID-19 model are acknowledged using a novel advanced intelligent computing infrastructure that joins artificial neural networks with unsupervised learning-based optimizers i.e., Genetic Algorithms (GA) and sequential quadratic programming (SQP). Unsupervised learning strategy is provided which depends on the wavelet basis's sequential deconstruction of stochastic data. The weights or selection values of neural networks are utilizing cumulative algorithms of Meyer wavelet artificial neural networks (MWANNs) optimized with global search Genetic Algorithms (GAs) and Sequential Quadratic Programming (SQP), referred to as MWANNs-GA-SQP and the design technique is utilized to determine the COVID-19 model for five different scenarios employing different step sizes and input intervals. The findings of this research article examined that in order to minimize the total disease transmission at the lowest cost and complexity, safety, focused medical care, and exterior sterilization methods applicability. The provided data is validated through various graphical simulations, which surely authenticate the effectiveness and robustness of the proposed solver. The suggested solver, MWANNs-GA-SQP, is tested in a variety of circumstances to examine that how reliable, safe, and tolerant. Using the proposed MWANNs hubristic intelligent approach, an objective optimization function is created in feed forward neural networking to minimize the mean square error. An investigation of the hybrid GA-SQP is used to confirm the accuracy and dependability of the MWANNs model results. Mean absolute graphs have been constructed to assess the integrity and efficiency of the proposed methodology. The accuracy and reliability of the suggested method are demonstrated by constantly achieving maximum variables of analytical assessment criteria computed for a large appropriate variety of distinct trials.
Collapse
Affiliation(s)
- Kottakkaran Sooppy Nisar
- Department of Mathematics, College of Science and Humanities in Al Kharj, Prince Sattam bin Abdulaziz University, 11942, Saudi Arabia; Saveetha School of Engineering, SIMATS, Chennai, India.
| | - Iqra Naz
- Department of Mathematics, COMSATS University Islamabad, Attock Campus, Pakistan.
| | - Muhammad Asif Zahoor Raja
- Future Technology Research Center, National Yunlin University of Science and Technology, 123 University Road, Section.3, Douliou, Yunlin 64002, Taiwan.
| | | |
Collapse
|
16
|
Zhang Z, Zhou L, Liu Q, Zheng Y, Tan X, Huang Z, Guo M, Wang X, Chen X, Liang S, Li W, Song K, Yan K, Li J, Li Q, Zhang Y, Yang S, Cai Z, Dai M, Xian Q, Shi ZL, Xu K, Lan K, Chen Y. The lethal K18-hACE2 knock-in mouse model mimicking the severe pneumonia of COVID-19 is practicable for antiviral development. Emerg Microbes Infect 2024; 13:2353302. [PMID: 38753462 PMCID: PMC11132709 DOI: 10.1080/22221751.2024.2353302] [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: 01/09/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
Animal models of COVID-19 facilitate the development of vaccines and antivirals against SARS-CoV-2. The efficacy of antivirals or vaccines may differ in different animal models with varied degrees of disease. Here, we introduce a mouse model expressing human angiotensin-converting enzyme 2 (ACE2). In this model, ACE2 with the human cytokeratin 18 promoter was knocked into the Hipp11 locus of C57BL/6J mouse by CRISPR - Cas9 (K18-hACE2 KI). Upon intranasal inoculation with high (3 × 105 PFU) or low (2.5 × 102 PFU) dose of SARS-CoV-2 wildtype (WT), Delta, Omicron BA.1, or Omicron BA.2 variants, all mice showed obvious infection symptoms, including weight loss, high viral loads in the lung, and interstitial pneumonia. 100% lethality was observed in K18-hACE2 KI mice infected by variants with a delay of endpoint for Delta and BA.1, and a significantly attenuated pathogenicity was observed for BA.2. The pneumonia of infected mice was accompanied by the infiltration of neutrophils and pulmonary fibrosis in the lung. Compared with K18-hACE2 Tg mice and HFH4-hACE2 Tg mice, K18-hACE2 KI mice are more susceptible to SARS-CoV-2. In the antivirals test, REGN10933 and Remdesivir had limited antiviral efficacies in K18-hACE2 KI mice upon the challenge of SARS-CoV-2 infections, while Nirmatrelvir, monoclonal antibody 4G4, and mRNA vaccines potently protected the mice from death. Our results suggest that the K18-hACE2 KI mouse model is lethal and stable for SARS-CoV-2 infection, and is practicable and stringent to antiviral development.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Li Zhou
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Qianyun Liu
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Yucheng Zheng
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Xue Tan
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Zhixiang Huang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Ming Guo
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Xin Wang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Xianying Chen
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Simeng Liang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Wenkang Li
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Kun Song
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Kun Yan
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Jiali Li
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Qiaohong Li
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Yuzhen Zhang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Shimin Yang
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
| | - Zeng Cai
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Ming Dai
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Qiaoyang Xian
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Zheng-Li Shi
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Ke Lan
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| | - Yu Chen
- State Key Laboratory of Virology, Modern Virology Research Center and RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, People’s Republic of China
- Institute for Vaccine Research, Animal Bio-Safety Level III Laboratory / Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, People’s Republic of China
| |
Collapse
|
17
|
Handrejk K, Schmitz KS, Veldhuis Kroeze EJB, van Dijk LLA, van Run P, Haagmans B, Moscona A, Porotto M, de Swart RL, de Vries RD, Rissmann M. Characterization of a SARS-CoV-2 Omicron BA.5 direct-contact transmission model in hamsters. NPJ VIRUSES 2024; 2:52. [PMID: 39512864 PMCID: PMC11537969 DOI: 10.1038/s44298-024-00061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024]
Abstract
As SARS-CoV-2 continues to evolve antigenically to escape vaccine- or infection-induced immunity, suitable animal models are needed to study novel interventions against viral variants. Syrian hamsters are often used because of their high susceptibility to SARS-CoV-2 and associated tissue damage in the respiratory tract. Here, we established a direct-contact transmission model for SARS-CoV-2 Omicron BA.5 in hamsters. First, we determined whether 103 or 104 TCID50 in a low-volume inoculum led to reproducible infection and viral shedding in male and female hamsters. Next, we determined the optimal co-housing timing and duration between donor and recipient hamsters required for consistent direct-contact transmission. Finally, we compared viral loads and histopathological lesions in the respiratory tissues of donor and recipient hamsters. Intranasal inoculation of hamsters with 103 TCID50 and 104 TCID50 Omicron BA.5 in 10 µl per nostril led to reproducible infection. Viral loads in the throat measured by RT-qPCR were comparable between male and female hamsters. Notably, the shedding of infectious virus was significantly higher in male hamsters. Compared to SARS-CoV-2 D614G, Omicron BA.5 infection reached lower viral loads, had a delayed peak of virus replication, and induced limited body weight loss. To ensure consistent direct-contact transmission from inoculated donor hamsters to naïve recipients, a co-housing duration of 24 h starting 20 h post-infection of the donors was optimal. We detected mild inflammation in the respiratory tract of donor and recipient hamsters, and viral loads were higher and peaked earlier in donor hamsters compared to recipient hamsters. Taken together, we developed a robust Omicron BA.5 direct-contact transmission model in hamsters, that provides a valuable tool to study novel interventions.
Collapse
Affiliation(s)
- Kim Handrejk
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Bart Haagmans
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Anne Moscona
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
| | - Matteo Porotto
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Melanie Rissmann
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
18
|
Ma J, Wu C, Xu J. The Development of Lung Tissue Engineering: From Biomaterials to Multicellular Systems. Adv Healthc Mater 2024; 13:e2401025. [PMID: 39206615 DOI: 10.1002/adhm.202401025] [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: 03/22/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
The challenge of the treatment of end-stage lung disease poses an urgent clinical demand for lung tissue engineering. Over the past few years, various lung tissue-engineered constructs are developed for lung tissue regeneration and respiratory pathology study. In this review, an overview of recent achievements in the field of lung tissue engineering is proposed. The introduction of lung structure and lung injury are stated briefly at first. After that, the lung tissue-engineered constructs are categorized into three types: acellular, monocellular, and multicellular systems. The different bioengineered constructs included in each system that can be applied to the reconstruction of the trachea, airway epithelium, alveoli, and even whole lung are described in detail, followed by the highlight of relevant representative research. Finally, the challenges and future directions of biomaterials, manufacturing technologies, and cells involved in lung tissue engineering are discussed. Overall, this review can provide referable ideas for the realization of functional lung regeneration and permanent lung substitution.
Collapse
Affiliation(s)
- Jingge Ma
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinfu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| |
Collapse
|
19
|
Creppy JR, Delache B, Lemaitre J, Horvat B, Vecellio L, Ducancel F. Administration of airborne pathogens in non-human primates. Inhal Toxicol 2024; 36:475-500. [PMID: 39388247 DOI: 10.1080/08958378.2024.2412685] [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: 10/16/2023] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Airborne pathogen scan penetrate in human respiratory tract and can cause illness. The use of animal models to predict aerosol deposition and study respiratory disease pathophysiology is therefore important for research and a prerequisite to test and study the mechanism of action of treatment. NHPs are relevant animal species for inhalation studies because of their similarities with humans in terms of anatomical structure, respiratory parameters and immune system. MATERIALS AND METHODS The aim of this review is to provide an overview of the state of the art of pathogen aerosol studies performed in non-human primates (NHPs). Herein, we present and discuss the deposition of aerosolized bacteria and viruses. In this review, we present important advantages of using NHPs as model for inhalation studies. RESULTS We demonstrate that deposition in the respiratory tract is not only a function of aerosol size but also the technique of administration influences the biological activity and site of aerosol deposition. Finally, we observe an influence of a region of pathogen deposition in the respiratory tract on the development of the pathophysiological effect in NHPs. CONCLUSION The wide range of methods used for the delivery of pathogento NHP respiratory airways is associated with varying doses and deposition profiles in the airways.
Collapse
Affiliation(s)
- Justina R Creppy
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Benoit Delache
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université de Lyon, Lyon, France
| | - Laurent Vecellio
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Frédéric Ducancel
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
20
|
Methwani DA, Deosthale N, Khadakkar S, Dhote K, Harkare V. Study of Otorhinolaryngological Manifestations in Symptomatic COVID-19-Positive Patients at Tertiary Health Care Hospital: A Cross-sectional Study. Int Arch Otorhinolaryngol 2024; 28:e597-e602. [PMID: 39464359 PMCID: PMC11511268 DOI: 10.1055/s-0044-1786831] [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: 11/02/2023] [Accepted: 03/31/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19), also referred to as the 2019 novel coronavirus, is caused by a single-stranded positive-sense RNA virus. This infectious agent spreads through respiratory routes, primarily utilizing aerosols. In our study, we shed light on ear, nose, and throat (ENT) manifestations, which can be considered as prognostic clinical biomarkers to reduce forthcoming complications among such critically ill patients. This makes it feasible for researchers to investigate or fetch early diagnosis in such cases with ease without the use of large, extensive hospital-base setups. Objective To study the otorhinolaryngological (ENT) manifestations in symptomatic COVID-19 patients. Methods From January to December 2021, a cross-sectional observational study was conducted at the Department of ENT of a tertiary care hospital in central India. All eligible symptomatic COVID-19 patients admitted to this institution during the study period were consecutively enrolled. The Institutional Ethics Committee gave its permission for the project. Results Out of the total of 1,375 patients, 78% of the patients diagnosed with COVID exhibited symptoms related to the ENT, while the remaining 22% did not display any ENT manifestations. Anosmia (71.2%), sore throat (55.62%), headache (44.3%), and loss of taste (66.3%) were among the most common ENT symptoms. Conclusion The present study highlights ENT manifestations, which play a crucial role in the early diagnosis of COVID-19 patients, ensuring faster treatment and isolation for better containment of the disease. Symptomatic treatment has shown efficacy, but objective tests are needed to prevent overestimation, understand pathogenesis, and enhance treatment.
Collapse
Affiliation(s)
- Disha Amar Methwani
- Department of ENT, N. K. P. Salve Institute of Medical Sciences & Research Centre and Lata Mangeshkar Hospital, Nagpur, Maharashtra, India
| | - Nitin Deosthale
- Department of ENT, N. K. P. Salve Institute of Medical Sciences & Research Centre and Lata Mangeshkar Hospital, Nagpur, Maharashtra, India
| | - Sonali Khadakkar
- Department of ENT, N. K. P. Salve Institute of Medical Sciences & Research Centre and Lata Mangeshkar Hospital, Nagpur, Maharashtra, India
| | - Kanchan Dhote
- Department of ENT, N. K. P. Salve Institute of Medical Sciences & Research Centre and Lata Mangeshkar Hospital, Nagpur, Maharashtra, India
| | - Vivek Harkare
- Department of ENT, N. K. P. Salve Institute of Medical Sciences & Research Centre and Lata Mangeshkar Hospital, Nagpur, Maharashtra, India
| |
Collapse
|
21
|
Ardalan M, Cool K, Gaudreault NN, Bold D, Rojas C, Mannix A, Seetahal J, Richt JA, Pogranichniy RM. Bison, Elk, and Other Captive Wildlife Species Humoral Immune Responses against SARS-CoV-2. Animals (Basel) 2024; 14:2829. [PMID: 39409778 PMCID: PMC11475800 DOI: 10.3390/ani14192829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for COVID-19, has been found to infect various domestic and wild animal species. In this study, convenience serum samples from 575 bison, 180 elk, and 147 samples from various wildlife species collected between 2020 and 2023 from several regions in the United States were analyzed for the presence of SARS-CoV-2-specific antibodies. Two commercial ELISA assays based on the inhibition of the SARS-CoV-2 receptor-binding domain (sVNT) or the nucleocapsid protein (N-ELISA) of SARS-CoV-2 were used. Positive samples from the sVNT were additionally evaluated using a conventional virus neutralization test (VNT). Our results indicated that 1.2% of bison, 2.2% of elk, and 4.1% of the other wildlife species serum samples were seropositive in the sVNT, whereas 4.2% of bison, 3.3% of elk, and 1.4% of the other captive wildlife species serum samples tested positive by the N-ELISA. Among the sVNT serum samples, two samples from bison, one sample from elk, and five serum samples from other wildlife species (one cheetah, one gorilla, two lions, and one hippopotamus) had neutralizing antibody titers in the VNT, indicating these species are susceptible to SARS-CoV-2 infection. These findings highlight the importance of broad surveillance efforts for the effective monitoring of SARS-CoV-2 in non-human hosts.
Collapse
Affiliation(s)
- Mehrnaz Ardalan
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
| | - Konner Cool
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
| | - Catherine Rojas
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Anna Mannix
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Janine Seetahal
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
| | - Roman M. Pogranichniy
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (M.A.); (N.N.G.); (J.A.R.)
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
22
|
Huang CY, Su SB, Chen KT. Surveillance strategies for SARS-CoV-2 infections through one health approach. Heliyon 2024; 10:e37128. [PMID: 39286214 PMCID: PMC11403048 DOI: 10.1016/j.heliyon.2024.e37128] [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: 05/06/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Coronavirus disease-2019 (COVID-19), caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), is an emergent disease that threatens global health. Public health structures and economic activities have been disrupted globally by the COVID-19 pandemic. Over 556.3 million confirmed cases and 6.3 million deaths have been reported. However, the exact mechanism of its emergence in humans remains unclear. SARS-CoV-2 is believed to have a zoonotic origin, suggesting a spillover route from animals to humans, which is potentially facilitated by wildlife farming and trade. The COVID-19 pandemic highlighted the importance of the One Health approach in managing threats of zoonosis in the human-animal-environment interaction. Implementing vigilant surveillance programs by adopting the One Health concept at the interfaces between wildlife, livestock, and humans is the most pertinent, practical, and actionable strategy for preventing and preparing for future pandemics of zoonosis, such as COVID-19 infection. This review summarizes the updated evidence of CoV infections in humans and animals and provides an appropriate strategy for preventive measures focused on surveillance systems through an On Health approach.
Collapse
Affiliation(s)
- Chien-Yuan Huang
- Division of Occupational Medicine, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Shih-Bin Su
- Department of Occupational Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Kow-Tong Chen
- Department of Occupational Medicine, Tainan Municipal Hospital (managed by Show Chwan Medical Care Corporation), Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
23
|
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: 0] [Impact Index Per Article: 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
|
24
|
Yang Q, Kelkar A, Manicassamy B, Neelamegham S. Conserved role of spike S2 domain N-glycosylation across beta-coronavirus family. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611372. [PMID: 39282346 PMCID: PMC11398505 DOI: 10.1101/2024.09.05.611372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Besides acting as an immunological shield, the N-glycans of SARS-CoV-2 are also critical for viral life cycle. As the S2 subunit of spike is highly conserved across beta-coronaviruses, we determined the functional significance of the five 'stem N-glycans' located in S2 between N1098-N1194. Studies were performed with 31 Asn-to-Gln mutants, beta-coronavirus virus-like particles and single-cycle viral replicons. Deletions of stem N-glycans enhanced S1 shedding from trimeric spike, reduced ACE2 binding and abolished syncytia formation. When three or more N-glycans were deleted, spike expression on cell surface and incorporation into virions was both reduced. Viral entry function was progressively lost upon deleting the N1098 glycan in combination with additional glycosite modifications. In addition to SARS-CoV-2, deleting stem N-glycans in SARS-CoV and MERS-CoV spike also prevented viral entry into target cells. These data suggest multiple functional roles for the stem N-glycans, and evolutionarily conserved properties for these complex carbohydrates across human beta-coronaviruses.
Collapse
Affiliation(s)
- Qi Yang
- Chemical & Biological Engineering, State University of New York, Buffalo, NY 14260, USA
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY 14260, USA
| | - Anju Kelkar
- Chemical & Biological Engineering, State University of New York, Buffalo, NY 14260, USA
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY 14260, USA
| | - Balaji Manicassamy
- Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Sriram Neelamegham
- Chemical & Biological Engineering, State University of New York, Buffalo, NY 14260, USA
- Cell, Gene and Tissue Engineering Center, State University of New York, Buffalo, NY 14260, USA
- Biomedical Engineering, State University of New York, Buffalo, NY 14260, USA
- Medicine, State University of New York, Buffalo, NY 14260, USA
- Clinical & Translational Research Center, Buffalo, NY 14260, USA
| |
Collapse
|
25
|
Breugem TI, Riesebosch S, Schipper D, Mykytyn AZ, van den Doel P, Segalés J, Lamers MM, Haagmans BL. Resistance to SARS-CoV-2 infection in camelid nasal organoids is associated with lack of ACE2 expression. NPJ VIRUSES 2024; 2:42. [PMID: 40295820 PMCID: PMC11721443 DOI: 10.1038/s44298-024-00054-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/12/2024] [Indexed: 04/30/2025]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects a variety of animal species. Susceptibility to SARS-CoV-2 is primarily determined by the utilization of the viral receptor, ACE2. SARS-CoV-2 can utilize a broad range of animal ACE2 isoforms in vitro, including the ACE2 from various camelid species. However, experimental infection of these animals does not lead to productive infection or seroconversion. In this study, we investigate the susceptibility of camelids to SARS-CoV-2 using novel well-differentiated camelid nasal organoids. We show that camelid nasal organoids are highly susceptible to Middle East respiratory syndrome coronavirus (MERS-CoV) infection, but not to infection with different SARS-CoV-2 variants (614G, BA.1 or EG.5.1.1). All viruses efficiently infected human airway organoids. Immunohistochemistry analysis revealed the absence of ACE2 on camelid nasal organoids and dromedary camel upper respiratory tract. In contrast, DPP4 was expressed in both camelid nasal organoids and the camel upper respiratory tract, which correlates with MERS-CoV infection. This study indicates that the camelid upper respiratory tract lacks expression of ACE2, which is associated with resistance to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Tim I Breugem
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Samra Riesebosch
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
- Infection and Immunity, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Debby Schipper
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anna Z Mykytyn
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Petra van den Doel
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joaquim Segalés
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Bellaterra, Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, 08193, Bellaterra, Barcelona, Spain
| | - Mart M Lamers
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands
- Programme of Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Bart L Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
26
|
Kim J, Roh H, Moon S, Jeon C, Baek S, Cho W, Sim JY, Jeong U. Wireless breathable face mask sensor for spatiotemporal 2D respiration profiling and respiratory diagnosis. Biomaterials 2024; 309:122579. [PMID: 38670033 DOI: 10.1016/j.biomaterials.2024.122579] [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: 11/22/2023] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024]
Abstract
Owing to air pollution and the pandemic outbreak, the need for quantitative pulmonary monitoring has greatly increased. The COVID-19 outbreak has aroused attention for comfortable wireless monitoring of respiratory profiles and more real-time diagnosis of respiratory diseases. Although respiration sensors have been investigated extensively with single-pixel sensors, 2D respiration profiling with a pixelated array sensor has not been demonstrated for both exhaling and inhaling. Since the pixelated array sensor allowed for simultaneous profiling of the nasal breathing and oral breathing, it provides essential respiratory information such as breathing patterns, respiration habit, breathing disorders. In this study, we introduced an air-permeable, stretchable, and a pixelated pressure sensor that can be integrated into a commercial face mask. The mask sensor showed a strain-independent pressure-sensing performance, providing 2D pressure profiles for exhalation and inhalation. Real-time 2D respiration profiles could monitor various respiratory behaviors, such as oral/nasal breathing, clogged nose, out-of-breath, and coughing. Furthermore, they could detect respiratory diseases, such as rhinitis, sleep apnea, and pneumonia. The 2D respiratory profiling mask sensor is expected to be employed for remote respiration monitoring and timely patient treatment.
Collapse
Affiliation(s)
- Jaehyun Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Heesung Roh
- Department of Convergence IT Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Sungmin Moon
- Department of Materials Science and Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Cheonhoo Jeon
- School of Electronics and Electrical Engineering, Dankook University, Yongin, Gyeonggi, 16890, South Korea
| | - Seunggoo Baek
- Department of Materials Science and Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Woosung Cho
- Department of Materials Science and Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Jae-Yoon Sim
- Department of Electrical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea.
| | - Unyong Jeong
- Department of Materials Science and Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea.
| |
Collapse
|
27
|
Liang K, Barnett KC, Hsu M, Chou WC, Bais SS, Riebe K, Xie Y, Nguyen TT, Oguin TH, Vannella KM, Hewitt SM, Chertow DS, Blasi M, Sempowski GD, Karlsson A, Koller BH, Lenschow DJ, Randell SH, Ting JPY. Initiator cell death event induced by SARS-CoV-2 in the human airway epithelium. Sci Immunol 2024; 9:eadn0178. [PMID: 38996010 PMCID: PMC11970318 DOI: 10.1126/sciimmunol.adn0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/14/2024] [Indexed: 07/14/2024]
Abstract
Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.
Collapse
Affiliation(s)
- Kaixin Liang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Oral and Craniofacial Biomedicine Program, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine C. Barnett
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin Hsu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wei-chun Chou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sachendra S. Bais
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | - Yuying Xie
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Tuong Thien Nguyen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Kevin M. Vannella
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Stephen M. Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S. Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Present address: RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Amelia Karlsson
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Present address: RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Beverly H. Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deborah J. Lenschow
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Scott H. Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jenny P.-Y. Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Keating SM, Higgins BW. New technologies in therapeutic antibody development: The next frontier for treating infectious diseases. Antiviral Res 2024; 227:105902. [PMID: 38734210 DOI: 10.1016/j.antiviral.2024.105902] [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: 01/29/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Adaptive immunity to viral infections requires time to neutralize and clear viruses to resolve infection. Fast growing and pathogenic viruses are quickly established, are highly transmissible and cause significant disease burden making it difficult to mount effective responses, thereby prolonging infection. Antibody-based passive immunotherapies can provide initial protection during acute infection, assist in mounting an adaptive immune response, or provide protection for those who are immune suppressed or immune deficient. Historically, plasma-derived antibodies have demonstrated some success in treating diseases caused by viral pathogens; nonetheless, limitations in access to product and antibody titer reduce success of this treatment modality. Monoclonal antibodies (mAbs) have proven an effective alternative, as it is possible to manufacture highly potent and specific mAbs against viral targets on an industrial scale. As a result, innovative technologies to discover, engineer and manufacture specific and potent antibodies have become an essential part of the first line of treatment in pathogenic viral infections. However, a mAb targeting a specific epitope will allow escape variants to outgrow, causing new variant strains to become dominant and resistant to treatment with that mAb. Methods to mitigate escape have included combining mAbs into cocktails, creating bi-specific or antibody drug conjugates but these strategies have also been challenged by the potential development of escape mutations. New technologies in developing antibodies made as recombinant polyclonal drugs can integrate the strength of poly-specific antibody responses to prevent mutational escape, while also incorporating antibody engineering to prevent antibody dependent enhancement and direct adaptive immune responses.
Collapse
Affiliation(s)
- Sheila M Keating
- GigaGen, Inc. (A Grifols Company), 75 Shoreway Road, San Carlos, CA, 94070, USA.
| | | |
Collapse
|
29
|
Zhang Y, Xu F, Wang T, Han Z, Shang H, Han K, Zhu P, Gao S, Wang X, Xue Y, Huang C, Chen Y, Liu G. Shared genetics and causal association between plasma levels of SARS-CoV-2 entry receptor ACE2 and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14873. [PMID: 39056224 PMCID: PMC11273102 DOI: 10.1111/cns.14873] [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: 04/04/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the highest risk of COVID-19 infection, hospitalization, and mortality. However, it remains largely unclear about the link between AD and COVID-19 outcomes. ACE2 is an entry receptor for SARS-CoV-2. Circulating ACE2 is a novel biomarker of death and associated with COVID-19 outcomes. METHODS Here, we explored the shared genetics and causal association between AD and plasma ACE2 levels using large-scale genome-wide association study, gene expression, expression quantitative trait loci, and high-throughput plasma proteomic profiling datasets. RESULTS We found a significant causal effect of genetically increased circulating ACE2 on increased risk of AD. Cross-trait association analysis identified 19 shared genetic variants, and three variants rs3104412, rs2395166, and rs3135344 at chromosome 6p21.32 were associated with COVID-19 infection, hospitalization, and severity. We mapped 19 variants to 117 genes, which were significantly upregulated in lung, spleen, and small intestine, downregulated in brain tissues, and involved in immune system, immune disease, and infectious disease pathways. The plasma proteins corresponding to LST1, AGER, TNXB, and APOC1 were predominantly associated with COVID-19 infection, ventilation, and death. CONCLUSION Together, our findings suggest the shared genetics and causal association between AD and plasma ACE2 levels, which may partially explain the link between AD and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of PathologyThe Affiliated Hospital of Weifang Medical UniversityWeifangChina
| | - Fang Xu
- Department of Neurology, Xuanwu Hospital, National Center for Neurological DisordersCapital Medical UniversityBeijingChina
| | - Tao Wang
- Academy for Advanced Interdisciplinary StudiesPeking UniversityBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| | - Zhifa Han
- Center of Respiratory Medicine, China–Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory MedicineChinese Acadamy of Medical Sciences, National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Hong Shang
- Department of NeurologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Kevin Han
- Department of StatisticsStanford UniversityStanfordCaliforniaUSA
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xiaojie Wang
- Department of NeurologyShenzhen Qianhai Shekou Free Trade Zone HospitalShenzhenChina
| | - Yanli Xue
- School of Biomedical EngineeringCapital Medical UniversityBeijingChina
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao SARChina
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public HealthWannan Medical CollegeWuhuChina
- Institute of Chronic Disease Prevention and ControlWannan Medical CollegeWuhuChina
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Department of Epidemiology and Biostatistics, School of Public HealthWannan Medical CollegeWuhuChina
- Institute of Chronic Disease Prevention and ControlWannan Medical CollegeWuhuChina
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Taishan Vocational College of NursingTaianChina
- Brain HospitalShengli Oilfield Central HospitalDongyingChina
| |
Collapse
|
30
|
Kang BM, Kim D, Kim J, Baek K, Park S, Shin HE, Lee MH, Kim M, Kim S, Lee Y, Kwon HJ. Analysis of SARS-CoV-2 Mutations after Nirmatrelvir Treatment in a Lung Cancer Xenograft Mouse Model. Biomol Ther (Seoul) 2024; 32:481-491. [PMID: 38835145 PMCID: PMC11214963 DOI: 10.4062/biomolther.2023.195] [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: 11/06/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 06/06/2024] Open
Abstract
Paxlovid is the first approved oral treatment for coronavirus disease 2019 and includes nirmatrelvir, a protease inhibitor targeting the main protease (Mpro) of SARS-CoV-2, as one of the key components. While some specific mutations emerged in Mpro were revealed to significantly reduce viral susceptibility to nirmatrelvir in vitro, there is no report regarding resistance to nirmatrelvir in patients and animal models for SARS-CoV-2 infection yet. We recently developed xenograft tumors derived from Calu-3 cells in immunodeficient mice and demonstrated extended replication of SARS-CoV-2 in the tumors. In this study, we investigated the effect of nirmatrelvir administration on SARS-CoV-2 replication. Treatment with nirmatrelvir after virus infection significantly reduced the replication of the parental SARS-CoV-2 and SARS-CoV-2 Omicron at 5 days post-infection (dpi). However, the virus titers were completely recovered at the time points of 15 and 30 dpi. The virus genomes in the tumors at 30 dpi were analyzed to investigate whether nirmatrelvir-resistant mutant viruses had emerged during the extended replication of SARS-CoV-2. Various mutations in several genes including ORF1ab, ORF3a, ORF7a, ORF7b, ORF8, and N occurred in the SARS-CoV-2 genome; however, no mutations were induced in the Mpro sequence by a single round of nirmatrelvir treatment, and none were observed even after two rounds of treatment. The parental SARS-CoV-2 and its sublineage isolates showed similar IC50 values of nirmatrelvir in Vero E6 cells. Therefore, it is probable that inducing viral resistance to nirmatrelvir in vivo is challenging differently from in vitro passage.
Collapse
Affiliation(s)
- Bo Min Kang
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Dongbum Kim
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jinsoo Kim
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyeongbin Baek
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Ha-Eun Shin
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Myeong-Heon Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Minyoung Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Suyeon Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyung-Joo Kwon
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
31
|
Milich KM, Morse SS. The reverse zoonotic potential of SARS-CoV-2. Heliyon 2024; 10:e33040. [PMID: 38988520 PMCID: PMC11234007 DOI: 10.1016/j.heliyon.2024.e33040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
There has been considerable emphasis recently on the zoonotic origins of emerging infectious diseases in humans, including the SARS-CoV-2 pandemic; however, reverse zoonoses (infections transmitted from humans to other animals) have received less attention despite their potential importance. The effects can be devastating for the infected species and can also result in transmission of the pathogen back to human populations or other animals either in the original form or as a variant. Humans have transmitted SARS-CoV-2 to other animals, and the virus is able to circulate and evolve in those species. As global travel resumes, the potential of SARS-CoV-2 as a reverse zoonosis threatens humans and endangered species. Nonhuman primates are of particular concern given their susceptibility to human respiratory infections. Enforcing safety measures for all people working in and visiting wildlife areas, especially those with nonhuman primates, and increasing access to safety measures for people living near protected areas that are home to nonhuman primates will help mitigate reverse zoonotic transmission.
Collapse
Affiliation(s)
- Krista M. Milich
- Department of Anthropology, Washington University in St. Louis, 1 Brookings Dr., St. Louis, MO, 63130, United States
| | - Stephen S. Morse
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th St., NY, NY, 10032, United States
| |
Collapse
|
32
|
Hamlin RE, Blish CA. Challenges and opportunities in long COVID research. Immunity 2024; 57:1195-1214. [PMID: 38865966 PMCID: PMC11210969 DOI: 10.1016/j.immuni.2024.05.010] [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: 01/02/2024] [Revised: 04/19/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
Long COVID (LC) is a condition in which patients do not fully recover from the initial SARS-CoV-2 infection but rather have persistent or new symptoms for months to years following the infection. Ongoing research efforts are investigating the pathophysiologic mechanisms of LC and exploring preventative and therapeutic treatment approaches for patients. As a burgeoning area of investigation, LC research can be structured to be more inclusive, innovative, and effective. In this perspective, we highlight opportunities for patient engagement and diverse research expertise, as well as the challenges of developing definitions and reproducible studies. Our intention is to provide a foundation for collaboration and progress in understanding the biomarkers and mechanisms driving LC.
Collapse
Affiliation(s)
| | - Catherine A Blish
- Department of Medicine, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
33
|
Wu TTH, Travaglini KJ, Rustagi A, Xu D, Zhang Y, Andronov L, Jang S, Gillich A, Dehghannasiri R, Martínez-Colón GJ, Beck A, Liu DD, Wilk AJ, Morri M, Trope WL, Bierman R, Weissman IL, Shrager JB, Quake SR, Kuo CS, Salzman J, Moerner W, Kim PS, Blish CA, Krasnow MA. Interstitial macrophages are a focus of viral takeover and inflammation in COVID-19 initiation in human lung. J Exp Med 2024; 221:e20232192. [PMID: 38597954 PMCID: PMC11009983 DOI: 10.1084/jem.20232192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/09/2024] [Accepted: 03/04/2024] [Indexed: 04/11/2024] Open
Abstract
Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.
Collapse
Affiliation(s)
- Timothy Ting-Hsuan Wu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Kyle J. Travaglini
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Arjun Rustagi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Yue Zhang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Leonid Andronov
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - SoRi Jang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Astrid Gillich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Roozbeh Dehghannasiri
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanny J. Martínez-Colón
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aimee Beck
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Dan Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Aaron J. Wilk
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Winston L. Trope
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rob Bierman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph B. Shrager
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Stephen R. Quake
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Christin S. Kuo
- Department of Pediatrics, Pulmonary Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia Salzman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - W.E. Moerner
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Catherine A. Blish
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Mark A. Krasnow
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| |
Collapse
|
34
|
De Meyer A, Meuleman P. Preclinical animal models to evaluate therapeutic antiviral antibodies. Antiviral Res 2024; 225:105843. [PMID: 38548022 DOI: 10.1016/j.antiviral.2024.105843] [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: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Despite the availability of effective preventative vaccines and potent small-molecule antiviral drugs, effective non-toxic prophylactic and therapeutic measures are still lacking for many viruses. The use of monoclonal and polyclonal antibodies in an antiviral context could fill this gap and provide effective virus-specific medical interventions. In order to develop these therapeutic antibodies, preclinical animal models are of utmost importance. Due to the variability in viral pathogenesis, immunity and overall characteristics, the most representative animal model for human viral infection differs between virus species. Therefore, throughout the years researchers sought to find the ideal preclinical animal model for each virus. The most used animal models in preclinical research include rodents (mice, ferrets, …) and non-human primates (macaques, chimpanzee, ….). Currently, antibodies are tested for antiviral efficacy against a variety of viruses including different hepatitis viruses, human immunodeficiency virus (HIV), influenza viruses, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rabies virus. This review provides an overview of the current knowledge about the preclinical animal models that are used for the evaluation of therapeutic antibodies for the abovementioned viruses.
Collapse
Affiliation(s)
- Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
35
|
Gheban-Roșca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. Immunohistochemical and Morphometric Analysis of Lung Tissue in Fatal COVID-19. Diagnostics (Basel) 2024; 14:914. [PMID: 38732328 PMCID: PMC11082993 DOI: 10.3390/diagnostics14090914] [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: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The primary targets of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the lungs are type I pneumocytes, macrophages, and endothelial cells. We aimed to identify lung cells targeted by SARS-CoV-2 using viral nucleocapsid protein staining and morphometric features on patients with fatal COVID-19. We conducted a retrospective analysis of fifty-one autopsy cases of individuals who tested positive for SARS-CoV-2. Demographic and clinical information were collected from forensic reports, and lung tissue was examined for microscopic lesions and the presence of specific cell types. Half of the evaluated cohort were older than 71 years, and the majority were male (74.5%). In total, 24 patients presented diffuse alveolar damage (DAD), and 50.9% had comorbidities (56.9% obesity, 33.3% hypertension, 15.7% diabetes mellitus). Immunohistochemical analysis showed a similar pattern of infected macrophages, infected type I pneumocytes, and endothelial cells, regardless of the presence of DAD (p > 0.5). The immunohistochemical reactivity score (IRS) was predominantly moderate but without significant differences between patients with and without DAD (p = 0.633 IRS for type I pneumocytes, p = 0.773 IRS for macrophage, and p = 0.737 for IRS endothelium). The nucleus/cytoplasm ratio shows lower values in patients with DAD (median: 0.29 vs. 0.35), but the difference only reaches a tendency for statistical significance (p = 0.083). Our study confirms the presence of infected macrophages, type I pneumocytes, and endothelial cells with a similar pattern in patients with and without diffuse alveolar damage.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
- Clinical Hospital for Infectious Diseases, 400348 Cluj-Napoca, Romania
| | - Bogdan-Alexandru Gheban
- County Emergency Clinical Hospital, 400006 Cluj-Napoca, Romania
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Bogdan Pop
- The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
- Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Daniela-Cristina Mironescu
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Vasile Costel Siserman
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| | - Sorana D. Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| |
Collapse
|
36
|
Losada A, Izquierdo-Useros N, Aviles P, Vergara-Alert J, Latino I, Segalés J, Gonzalez SF, Cuevas C, Raïch-Regué D, Muñoz-Alonso MJ, Perez-Zsolt D, Muñoz-Basagoiti J, Rodon J, Chang LA, Warang P, Singh G, Brustolin M, Cantero G, Roca N, Pérez M, Bustos-Morán E, White K, Schotsaert M, García-Sastre A. Plitidepsin as an Immunomodulator against Respiratory Viral Infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1307-1318. [PMID: 38416036 PMCID: PMC10984758 DOI: 10.4049/jimmunol.2300426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Plitidepsin is a host-targeted compound known for inducing a strong anti-SARS-CoV-2 activity, as well as for having the capacity of reducing lung inflammation. Because IL-6 is one of the main cytokines involved in acute respiratory distress syndrome, the effect of plitidepsin in IL-6 secretion in different in vitro and in vivo experimental models was studied. A strong plitidepsin-mediated reduction of IL-6 was found in human monocyte-derived macrophages exposed to nonproductive SARS-CoV-2. In resiquimod (a ligand of TLR7/8)-stimulated THP1 human monocytes, plitidepsin-mediated reductions of IL-6 mRNA and IL-6 levels were also noticed. Additionally, although resiquimod-induced binding to DNA of NF-κB family members was unaffected by plitidepsin, a decrease in the regulated transcription by NF-κB (a key transcription factor involved in the inflammatory cascade) was observed. Furthermore, the phosphorylation of p65 that is required for full transcriptional NF-κB activity was significantly reduced by plitidepsin. Moreover, decreases of IL-6 levels and other proinflammatory cytokines were also seen in either SARS-CoV-2 or H1N1 influenza virus-infected mice, which were treated at low enough plitidepsin doses to not induce antiviral effects. In summary, plitidepsin is a promising therapeutic agent for the treatment of viral infections, not only because of its host-targeted antiviral effect, but also for its immunomodulatory effect, both of which were evidenced in vitro and in vivo by the decrease of proinflammatory cytokines.
Collapse
Affiliation(s)
- Alejandro Losada
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute, Can Ruti Campus, Badalona, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Aviles
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Júlia Vergara-Alert
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Irene Latino
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Joaquim Segalés
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Carmen Cuevas
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | | | - María J Muñoz-Alonso
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | | | | | - Jordi Rodon
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marco Brustolin
- Unit of Entomology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Guillermo Cantero
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Núria Roca
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mònica Pérez
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Eugenio Bustos-Morán
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Kris White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
37
|
Fang JY, Huang KY, Wang TH, Lin ZC, Chen CC, Chang SY, Chen EL, Chao TL, Yang SC, Yang PC, Chen CY. Development of nanoparticles incorporated with quercetin and ACE2-membrane as a novel therapy for COVID-19. J Nanobiotechnology 2024; 22:169. [PMID: 38609998 PMCID: PMC11015574 DOI: 10.1186/s12951-024-02435-2] [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: 10/15/2023] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION Angiotensin-converting enzyme 2 (ACE2) and AXL tyrosine kinase receptor are known to be involved in the SARS-CoV-2 entry of the host cell. Therefore, targeting ACE2 and AXL should be an effective strategy to inhibit virus entry into cells. However, developing agents that can simultaneously target ACE2 and AXL remains a formidable task. The natural compound quercetin has been shown to inhibit AXL expression. MATERIALS AND METHODS In this study, we employed PLGA nanoparticles to prepare nanoparticles encapsulated with quercetin, coated with ACE2-containing cell membranes, or encapsulated with quercetin and then coated with ACE-2-containing cell membranes. These nanoparticles were tested for their abilities to neutralize or inhibit viral infection. RESULTS Our data showed that nanoparticles encapsulated with quercetin and then coated with ACE2-containing cell membrane inhibited the expression of AXL without causing cytotoxic activity. Nanoparticles incorporated with both quercetin and ACE2-containing cell membrane were found to be able to neutralize pseudo virus infection and were more effective than free quercetin and nanoparticles encapsulated with quercetin at inhibition of pseudo virus and SARS-CoV-2 infection. CONCLUSIONS We have shown that the biomimetic nanoparticles incorporated with both ACE-2 membrane and quercetin showed the most antiviral activity and may be further explored for clinical application.
Collapse
Affiliation(s)
- Jia-You Fang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Yen Huang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- National Taiwan University YongLin Institute of Health, Taipei, Taiwan
- Graduate School of Advanced Technology (Program for Precision Health and Intelligent Medicine), National Taiwan University, Taipei, Taiwan
| | - Tong-Hong Wang
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Biobank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Chin-Chuan Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Biobank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - En-Li Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shuenn-Chen Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pan-Chyr Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
- , No.1, Sec 1, Jen-Ai Rd, R.O.C, 100225, Taipei, Taiwan.
| | - Chi-Yuan Chen
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Biobank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- , No.261, Wenhua 1st Rd., Guishan Dist, 33303, Taoyuan City, Taiwan.
| |
Collapse
|
38
|
Warner BM, Chan M, Tailor N, Vendramelli R, Audet J, Meilleur C, Truong T, Garnett L, Willman M, Soule G, Tierney K, Albietz A, Moffat E, Higgins R, Santry LA, Leacy A, Pham PH, Yates JGE, Pei Y, Safronetz D, Strong JE, Susta L, Embury-Hyatt C, Wootton SK, Kobasa D. Mucosal Vaccination with a Newcastle Disease Virus-Vectored Vaccine Reduces Viral Loads in SARS-CoV-2-Infected Cynomolgus Macaques. Vaccines (Basel) 2024; 12:404. [PMID: 38675786 PMCID: PMC11054841 DOI: 10.3390/vaccines12040404] [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: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged following an outbreak of unexplained viral illness in China in late 2019. Since then, it has spread globally causing a pandemic that has resulted in millions of deaths and has had enormous economic and social consequences. The emergence of SARS-CoV-2 saw the rapid and widespread development of a number of vaccine candidates worldwide, and this never-before-seen pace of vaccine development led to several candidates progressing immediately through clinical trials. Many countries have now approved vaccines for emergency use, with large-scale vaccination programs ongoing. Despite these successes, there remains a need for ongoing pre-clinical and clinical development of vaccine candidates against SARS-CoV-2, as well as vaccines that can elicit strong mucosal immune responses. Here, we report on the efficacy of a Newcastle disease virus-vectored vaccine candidate expressing SARS-CoV-2 spike protein (NDV-FLS) administered to cynomolgus macaques. Macaques given two doses of the vaccine via respiratory immunization developed robust immune responses and had reduced viral RNA levels in nasal swabs and in the lower airway. Our data indicate that NDV-FLS administered mucosally provides significant protection against SARS-CoV-2 infection, resulting in reduced viral burden and disease manifestation, and should be considered as a viable candidate for clinical development.
Collapse
Affiliation(s)
- Bryce M. Warner
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Mable Chan
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Nikesh Tailor
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Robert Vendramelli
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Jonathan Audet
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Courtney Meilleur
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Thang Truong
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Lauren Garnett
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Marnie Willman
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Geoff Soule
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Kevin Tierney
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Alixandra Albietz
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Estella Moffat
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3R2, Canada; (E.M.); (C.E.-H.)
| | - Rick Higgins
- Department of Radiology, Health Sciences Center, Winnipeg, MB R3A 1S1, Canada;
| | - Lisa A. Santry
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Alexander Leacy
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Phuc H. Pham
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Jacob G. E. Yates
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - David Safronetz
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - James E. Strong
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3R2, Canada; (E.M.); (C.E.-H.)
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Darwyn Kobasa
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
39
|
Dufloo J, Sanjuán R. Temperature impacts SARS-CoV-2 spike fusogenicity and evolution. mBio 2024; 15:e0336023. [PMID: 38411986 PMCID: PMC11005339 DOI: 10.1128/mbio.03360-23] [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: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
SARS-CoV-2 infects both the upper and lower respiratory tracts, which are characterized by different temperatures (33°C and 37°C, respectively). In addition, fever is a common COVID-19 symptom. SARS-CoV-2 has been shown to replicate more efficiently at low temperatures, but the effect of temperature on different viral proteins remains poorly understood. Here, we investigate how temperature affects the SARS-CoV-2 spike function and evolution. We first observed that increasing temperature from 33°C to 37°C or 39°C increased spike-mediated cell-cell fusion. We then experimentally evolved a recombinant vesicular stomatitis virus expressing the SARS-CoV-2 spike at these different temperatures. We found that spike-mediated cell-cell fusion was maintained during evolution at 39°C but was lost in a high proportion of viruses that evolved at 33°C or 37°C. Consistently, sequencing of the spikes evolved at 33°C or 37°C revealed the accumulation of mutations around the furin cleavage site, a region that determines cell-cell fusion, whereas this did not occur in spikes evolved at 39°C. Finally, using site-directed mutagenesis, we found that disruption of the furin cleavage site had a temperature-dependent effect on spike-induced cell-cell fusion and viral fitness. Our results suggest that variations in body temperature may affect the activity and diversification of the SARS-CoV-2 spike. IMPORTANCE When it infects humans, SARS-CoV-2 is exposed to different temperatures (e.g., replication site and fever). Temperature has been shown to strongly impact SARS-CoV-2 replication, but how it affects the activity and evolution of the spike protein remains poorly understood. Here, we first show that high temperatures increase the SARS-CoV-2 spike fusogenicity. Then, we demonstrate that the evolution of the spike activity and variants depends on temperature. Finally, we show that the functional effect of specific spike mutations is temperature-dependent. Overall, our results suggest that temperature may be a factor influencing the activity and adaptation of the SARS-CoV-2 spike in vivo, which will help understanding viral tropism, pathogenesis, and evolution.
Collapse
Affiliation(s)
- Jérémy Dufloo
- Institute for Integrative Systems Biology, Consejo Superior de Investigaciones Científicas-Universitat de València, Paterna, València, Spain
| | - Rafael Sanjuán
- Institute for Integrative Systems Biology, Consejo Superior de Investigaciones Científicas-Universitat de València, Paterna, València, Spain
| |
Collapse
|
40
|
Park D, Kim SM, Jang H, Kim K, Ji HY, Yang H, Kwon W, Kang Y, Hwang S, Kim H, Casel MAB, Choi I, Yang JS, Lee JY, Choi YK. Differential beta-coronavirus infection dynamics in human bronchial epithelial organoids. J Med Virol 2024; 96:e29600. [PMID: 38591240 DOI: 10.1002/jmv.29600] [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: 12/27/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
The lower respiratory system serves as the target and barrier for beta-coronavirus (beta-CoV) infections. In this study, we explored beta-CoV infection dynamics in human bronchial epithelial (HBE) organoids, focusing on HCoV-OC43, SARS-CoV, MERS-CoV, and SARS-CoV-2. Utilizing advanced organoid culture techniques, we observed robust replication for all beta-CoVs, particularly noting that SARS-CoV-2 reached peak viral RNA levels at 72 h postinfection. Through comprehensive transcriptomic analysis, we identified significant shifts in cell population dynamics, marked by an increase in goblet cells and a concurrent decrease in ciliated cells. Furthermore, our cell tropism analysis unveiled distinct preferences in viral targeting: HCoV-OC43 predominantly infected club cells, while SARS-CoV had a dual tropism for goblet and ciliated cells. In contrast, SARS-CoV-2 primarily infected ciliated cells, and MERS-CoV showed a marked affinity for goblet cells. Host factor analysis revealed the upregulation of genes encoding viral receptors and proteases. Notably, HCoV-OC43 induced the unfolded protein response pathway, which may facilitate viral replication. Our study also reveals a complex interplay between inflammatory pathways and the suppression of interferon responses during beta-CoV infections. These findings provide insights into host-virus interactions and antiviral defense mechanisms, contributing to our understanding of beta-CoV infections in the respiratory tract.
Collapse
Affiliation(s)
- Dongbin Park
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Se-Mi Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Hobin Jang
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Kanghee Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Ho Young Ji
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Heedong Yang
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Woohyun Kwon
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Yeonglim Kang
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Suhee Hwang
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Hyunjoon Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Mark Anthony B Casel
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Issac Choi
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jeong-Sun Yang
- Division of Viral Diseases, Center for Laboratory Control of Infectious Disease, Korea National Institute of Health (KNIH), Cheongju, Republic of Korea
| | - Joo-Yeon Lee
- Division of Viral Diseases, Center for Laboratory Control of Infectious Disease, Korea National Institute of Health (KNIH), Cheongju, Republic of Korea
| | - Young Ki Choi
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
41
|
Prete A, Matrone A, Plebani R. State of the Art in 3D Culture Models Applied to Thyroid Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:520. [PMID: 38674166 PMCID: PMC11051914 DOI: 10.3390/medicina60040520] [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: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/28/2024]
Abstract
Thyroid cancer (TC) is the prevalent endocrine tumor with a rising incidence, particularly in higher-income countries, leading to an increased interest in its management and treatment. While overall, survival rates for TC are usually favorable, advanced cases, especially with metastasis and specific histotypes, pose challenges with poorer outcomes, advocating the need of systemic treatments. Targeted therapies have shown efficacy in both preclinical models and clinical trials but face issues of resistance, since they usually induce partial and transient response. These resistance phenomena are currently only partially addressed by traditional preclinical models. This review explores the limitations of traditional preclinical models and emphasizes the potential of three-dimensional (3D) models, such as transwell assays, spheroids, organoids, and organ-on-chip technology in providing a more comprehensive understanding of TC pathogenesis and treatment responses. We reviewed their use in the TC field, highlighting how they can produce new interesting insights. Finally, the advent of organ-on-chip technology is currently revolutionizing preclinical research, offering dynamic, multi-cellular systems that replicate the complexity of human organs and cancer-host interactions.
Collapse
Affiliation(s)
- Alessandro Prete
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University of Pisa, 56122 Pisa, Italy;
| | - Antonio Matrone
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University of Pisa, 56122 Pisa, Italy;
| | - Roberto Plebani
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University, 66100 Chieti-Pescara, Italy;
| |
Collapse
|
42
|
Qin L, Sun Y, Gao N, Ling G, Zhang P. Nanotechnology of inhalable vaccines for enhancing mucosal immunity. Drug Deliv Transl Res 2024; 14:597-620. [PMID: 37747597 DOI: 10.1007/s13346-023-01431-7] [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] [Accepted: 09/05/2023] [Indexed: 09/26/2023]
Abstract
Vaccines are the cornerstone of world health. The majority of vaccines are formulated as injectable products, facing the drawbacks of cold chain transportation, needle-stick injuries, and primary systemic immunity. Inhalable vaccines exhibited unique advantages due to their small dose, easy to use, quick effect, and simultaneous induction of mucosal and systemic responses. Facing global pandemics, especially the coronavirus disease 2019 (COVID-19), a majority of inhalable vaccines are in preclinical or clinical trials. A better understanding of advanced delivery technologies of inhalable vaccines may provide new scientific insights for developing inhalable vaccines. In this review article, detailed immune mechanisms involving mucosal, cellular, and humoral immunity were described. The preparation methods of inhalable vaccines were then introduced. Advanced nanotechnologies of inhalable vaccines containing inhalable nucleic acid vaccines, inhalable adenovirus vector vaccines, novel adjuvant-assisted inhalable vaccines, and biomaterials for inhalable vaccine delivery were emphatically discussed. Meanwhile, the latest clinical progress in inhalable vaccines for COVID-19 and tuberculosis was discussed.
Collapse
Affiliation(s)
- Li Qin
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., No. 243, Gongyebei Road, Jinan, 250100, China
| | - Nan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
43
|
Zhang J, Liu Y. Epithelial stem cells and niches in lung alveolar regeneration and diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:17-26. [PMID: 38645714 PMCID: PMC11027191 DOI: 10.1016/j.pccm.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Indexed: 04/23/2024]
Abstract
Alveoli serve as the functional units of the lungs, responsible for the critical task of blood-gas exchange. Comprising type I (AT1) and type II (AT2) cells, the alveolar epithelium is continuously subject to external aggressors like pathogens and airborne particles. As such, preserving lung function requires both the homeostatic renewal and reparative regeneration of this epithelial layer. Dysfunctions in these processes contribute to various lung diseases. Recent research has pinpointed specific cell subgroups that act as potential stem or progenitor cells for the alveolar epithelium during both homeostasis and regeneration. Additionally, endothelial cells, fibroblasts, and immune cells synergistically establish a nurturing microenvironment-or "niche"-that modulates these epithelial stem cells. This review aims to consolidate the latest findings on the identities of these stem cells and the components of their niche, as well as the molecular mechanisms that govern them. Additionally, this article highlights diseases that arise due to perturbations in stem cell-niche interactions. We also discuss recent technical innovations that have catalyzed these discoveries. Specifically, this review underscores the heterogeneity, plasticity, and dynamic regulation of these stem cell-niche systems. It is our aspiration that a deeper understanding of the fundamental cellular and molecular mechanisms underlying alveolar homeostasis and regeneration will open avenues for identifying novel therapeutic targets for conditions such as chronic obstructive pulmonary disease (COPD), fibrosis, coronavirus disease 2019 (COVID-19), and lung cancer.
Collapse
Affiliation(s)
- Jilei Zhang
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yuru Liu
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
44
|
Ma L, Zhu H, Jiang Y, Kong X, Gao P, Liu Y, Zhao M, Deng G, Cao Y. Development of a Novel Multiplex PCR Method for the Rapid Detection of SARS-CoV-2, Influenza A Virus, and Influenza B Virus. Int J Anal Chem 2024; 2024:4950391. [PMID: 38456096 PMCID: PMC10919977 DOI: 10.1155/2024/4950391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 02/03/2024] [Accepted: 02/10/2024] [Indexed: 03/09/2024] Open
Abstract
Objective A sensitive and specific multiplex fluorescence rapid detection method was established for simultaneous detection of SARS-CoV-2, influenza A virus, and influenza B virus in a self-made device within 30 min, with a minimum detection limit of 200 copies/mL. Methods Based on the genome sequences of SARS-CoV-2, influenza A virus (FluA), and influenza B virus (FluB) with reference to the Chinese Center for Disease Control and Prevention and related literature, specific primers were designed, and a multiplex fluorescent PCR system was established. The simultaneous and rapid detection of SARS-CoV-2, FluA, and FluB was achieved by optimizing the concentrations of Taq DNA polymerase as well as primers, probes, and Mg2+. The minimum detection limits of the nucleic acid rapid detection system for SARS-CoV-2, FluA, and FluB were evaluated. Results By optimizing the amplification system, the N enzyme with the best amplification performance was selected, and the optimal concentration of Mg2+ in the multiamplification system was 3 mmol/L; the final concentrations of SARS-CoV-2 NP probe and primer were 0.15 μmol/L and 0.2 μmol/L, respectively; the final concentrations of SARS-CoV-2 ORF probe and primer were both 0.15 μmol/L; the final concentrations of FluA probe and primer were 0.2 μmol/L and 0.3 μmol/L, respectively; the final concentrations of FluB probe and primer were 0.15 μmol/L and 0.25 μmol/L, respectively. Conclusion A multiplex real-time quantitative fluorescence RT-PCR system for three respiratory viruses of SARS-CoV-2, FluA, and FluB was established with a high amplification efficiency and sensitivity reaching 200 copies/mL for all samples. Combined with the automated microfluidic nucleic acid detection system, the system can achieve rapid detection in 30 minutes.
Collapse
Affiliation(s)
- Liang Ma
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Haoyan Zhu
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yongwei Jiang
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiaomu Kong
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Peng Gao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi Liu
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Meimei Zhao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Guoxiong Deng
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
45
|
Nederlof RA, de la Garza MA, Bakker J. Perspectives on SARS-CoV-2 Cases in Zoological Institutions. Vet Sci 2024; 11:78. [PMID: 38393096 PMCID: PMC10893009 DOI: 10.3390/vetsci11020078] [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: 01/06/2024] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in a zoological institution were initially reported in March 2020. Since then, at least 94 peer-reviewed cases have been reported in zoos worldwide. Among the affected animals, nonhuman primates, carnivores, and artiodactyls appear to be most susceptible to infection, with the Felidae family accounting for the largest number of reported cases. Clinical symptoms tend to be mild across taxa; although, certain species exhibit increased susceptibility to disease. A variety of diagnostic tools are available, allowing for initial diagnostics and for the monitoring of infectious risk. Whilst supportive therapy proves sufficient in most cases, monoclonal antibody therapy has emerged as a promising additional treatment option. Effective transmission of SARS-CoV-2 in some species raises concerns over potential spillover and the formation of reservoirs. The occurrence of SARS-CoV-2 in a variety of animal species may contribute to the emergence of variants of concern due to altered viral evolutionary constraints. Consequently, this review emphasizes the need for effective biosecurity measures and surveillance strategies to prevent and control SARS-CoV-2 infections in zoological institutions.
Collapse
Affiliation(s)
| | - Melissa A. de la Garza
- Michale E. Keeling Center for Comparative Medicine and Research, University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Jaco Bakker
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
46
|
Hu B, Guo H, Si H, Shi Z. Emergence of SARS and COVID-19 and preparedness for the next emerging disease X. Front Med 2024; 18:1-18. [PMID: 38561562 DOI: 10.1007/s11684-024-1066-6] [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: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 04/04/2024]
Abstract
Severe acute respiratory syndrome (SARS) and Coronavirus disease 2019 (COVID-19) are two human Coronavirus diseases emerging in this century, posing tremendous threats to public health and causing great loss to lives and economy. In this review, we retrospect the studies tracing the molecular evolution of SARS-CoV, and we sort out current research findings about the potential ancestor of SARS-CoV-2. Updated knowledge about SARS-CoV-2-like viruses found in wildlife, the animal susceptibility to SARS-CoV-2, as well as the interspecies transmission risk of SARS-related coronaviruses (SARSr-CoVs) are gathered here. Finally, we discuss the strategies of how to be prepared against future outbreaks of emerging or re-emerging coronaviruses.
Collapse
Affiliation(s)
- Ben Hu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hua Guo
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Haorui Si
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhengli Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
47
|
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
|
48
|
Zhang J, Rissmann M, Kuiken T, Haagmans BL. Comparative Pathogenesis of Severe Acute Respiratory Syndrome Coronaviruses. ANNUAL REVIEW OF PATHOLOGY 2024; 19:423-451. [PMID: 37832946 DOI: 10.1146/annurev-pathol-052620-121224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Over the last two decades the world has witnessed the global spread of two genetically related highly pathogenic coronaviruses, severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. However, the impact of these outbreaks differed significantly with respect to the hospitalizations and fatalities seen worldwide. While many studies have been performed recently on SARS-CoV-2, a comparative pathogenesis analysis with SARS-CoV may further provide critical insights into the mechanisms of disease that drive coronavirus-induced respiratory disease. In this review, we comprehensively describe clinical and experimental observations related to transmission and pathogenesis of SARS-CoV-2 in comparison with SARS-CoV, focusing on human, animal, and in vitro studies. By deciphering the similarities and disparities of SARS-CoV and SARS-CoV-2, in terms of transmission and pathogenesis mechanisms, we offer insights into the divergent characteristics of these two viruses. This information may also be relevant to assessing potential novel introductions of genetically related highly pathogenic coronaviruses.
Collapse
Affiliation(s)
- Jingshu Zhang
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands;
| | - Melanie Rissmann
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands;
| | - Thijs Kuiken
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands;
| | - Bart L Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands;
| |
Collapse
|
49
|
Zhuang J, Yan Z, Zhou T, Li Y, Wang H. The role of receptors in the cross-species spread of coronaviruses infecting humans and pigs. Arch Virol 2024; 169:35. [PMID: 38265497 DOI: 10.1007/s00705-023-05956-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/19/2023] [Indexed: 01/25/2024]
Abstract
The pandemic caused by SARS-CoV-2, which has proven capable of infecting over 30 animal species, highlights the critical need for understanding the mechanisms of cross-species transmission and the emergence of novel coronavirus strains. The recent discovery of CCoV-HuPn-2018, a recombinant alphacoronavirus from canines and felines that can infect humans, along with evidence of SARS-CoV-2 infection in pig cells, underscores the potential for coronaviruses to overcome species barriers. This review investigates the origins and cross-species transmission of both human and porcine coronaviruses, with a specific emphasis on the instrumental role receptors play in this process.
Collapse
Affiliation(s)
- Jie Zhuang
- Department of Basic Veterinary Medicine, College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhiwei Yan
- Department of Basic Veterinary Medicine, College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000, China
| | - Tiezhong Zhou
- Department of Basic Veterinary Medicine, College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000, China
| | - Yonggang Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Huinuan Wang
- Department of Basic Veterinary Medicine, College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
50
|
Berry N, Mee ET, Almond N, Rose NJ. The Impact and Effects of Host Immunogenetics on Infectious Disease Studies Using Non-Human Primates in Biomedical Research. Microorganisms 2024; 12:155. [PMID: 38257982 PMCID: PMC10818626 DOI: 10.3390/microorganisms12010155] [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: 09/13/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Understanding infectious disease pathogenesis and evaluating novel candidate treatment interventions for human use frequently requires prior or parallel analysis in animal model systems. While rodent species are frequently applied in such studies, there are situations where non-human primate (NHP) species are advantageous or required. These include studies of animals that are anatomically more akin to humans, where there is a need to interrogate the complexity of more advanced biological systems or simply reflect susceptibility to a specific infectious agent. The contribution of different arms of the immune response may be addressed in a variety of NHP species or subspecies in specific physiological compartments. Such studies provide insights into immune repertoires not always possible from human studies. However, genetic variation in outbred NHP models may confound, or significantly impact the outcome of a particular study. Thus, host factors need to be considered when undertaking such studies. Considerable knowledge of the impact of host immunogenetics on infection dynamics was elucidated from HIV/SIV research. NHP models are now important for studies of emerging infections. They have contributed to delineating the pathogenesis of SARS-CoV-2/COVID-19, which identified differences in outcomes attributable to the selected NHP host. Moreover, their use was crucial in evaluating the immunogenicity and efficacy of vaccines against COVID-19 and establishing putative correlates of vaccine protection. More broadly, neglected or highly pathogenic emerging or re-emergent viruses may be studied in selected NHPs. These studies characterise protective immune responses following infection or the administration of candidate immunogens which may be central to the accelerated licensing of new vaccines. Here, we review selected aspects of host immunogenetics, specifically MHC background and TRIM5 polymorphism as exemplars of adaptive and innate immunity, in commonly used Old and New World host species. Understanding this variation within and between NHP species will ensure that this valuable laboratory source is used most effectively to combat established and emerging virus infections and improve human health worldwide.
Collapse
Affiliation(s)
- Neil Berry
- Research & Development—Science, Research and Innovation, Medicines and Healthcare products Regulatory Agency, South Mimms, Hertfordshire EN6 3QG, UK; (E.T.M.); (N.A.); (N.J.R.)
| | | | | | | |
Collapse
|