201
|
Menéndez SG, Martín Giménez VM, Holick MF, Barrantes FJ, Manucha W. COVID-19 and neurological sequelae: Vitamin D as a possible neuroprotective and/or neuroreparative agent. Life Sci 2022; 297:120464. [PMID: 35271880 PMCID: PMC8898786 DOI: 10.1016/j.lfs.2022.120464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 12/16/2022]
Abstract
SARS-CoV-2, the etiological agent of the current COVID-19 pandemic, belongs to a broad family of coronaviruses that also affect humans. SARS-CoV-2 infection usually leads to bilateral atypical pneumonia with significant impairment of respiratory function. However, the infectious capacity of SARS-CoV-2 is not limited to the respiratory system, but may also affect other vital organs such as the brain. The central nervous system is vulnerable to cell damage via direct invasion or indirect virus-related effects leading to a neuroinflammatory response, processes possibly associated with a decrease in the activity of angiotensin II converting enzyme (ACE2), the canonical cell-surface receptor for SARS-CoV-2. This enzyme regulates neuroprotective and neuroimmunomodulatory functions and can neutralize both inflammation and oxidative stress generated at the cellular level. Furthermore, there is evidence of an association between vitamin D deficiency and predisposition to the development of severe forms of COVID-19, with its possible neurological and neuropsychiatric sequelae: vitamin D has the ability to down-modulate the effects of neuroinflammatory cytokines, among other anti-inflammatory/immunomodulatory effects, thus attenuating harmful consequences of COVID-19. This review critically analyzes current evidence supporting the notion that vitamin D may act as a neuroprotective and neuroreparative agent against the neurological sequelae of COVID-19.
Collapse
Affiliation(s)
- Sebastián García Menéndez
- Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina
| | - Virna Margarita Martín Giménez
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, San Juan, Argentina
| | - Michael F. Holick
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Medical Campus, 715 Albany St #437, Boston, MA 02118, USA
| | - Francisco J. Barrantes
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina
| | - Walter Manucha
- Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina,Corresponding author at: Pharmacology Area, Pathology Department, Medical Sciences College, National University of Cuyo, Mendoza CP5500, Argentina
| |
Collapse
|
202
|
Baselga M, Güemes A, Alba JJ, Schuhmacher AJ. SARS-CoV-2 Droplet and Airborne Transmission Heterogeneity. J Clin Med 2022; 11:2607. [PMID: 35566733 PMCID: PMC9099777 DOI: 10.3390/jcm11092607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/21/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022] Open
Abstract
The spread dynamics of the SARS-CoV-2 virus have not yet been fully understood after two years of the pandemic. The virus's global spread represented a unique scenario for advancing infectious disease research. Consequently, mechanistic epidemiological theories were quickly dismissed, and more attention was paid to other approaches that considered heterogeneity in the spread. One of the most critical advances in aerial pathogens transmission was the global acceptance of the airborne model, where the airway is presented as the epicenter of the spread of the disease. Although the aerodynamics and persistence of the SARS-CoV-2 virus in the air have been extensively studied, the actual probability of contagion is still unknown. In this work, the individual heterogeneity in the transmission of 22 patients infected with COVID-19 was analyzed by close contact (cough samples) and air (environmental samples). Viral RNA was detected in 2/19 cough samples from patient subgroups, with a mean Ct (Cycle Threshold in Quantitative Polymerase Chain Reaction analysis) of 25.7 ± 7.0. Nevertheless, viral RNA was only detected in air samples from 1/8 patients, with an average Ct of 25.0 ± 4.0. Viral load in cough samples ranged from 7.3 × 105 to 8.7 × 108 copies/mL among patients, while concentrations between 1.1-4.8 copies/m3 were found in air, consistent with other reports in the literature. In patients undergoing follow-up, no viral load was found (neither in coughs nor in the air) after the third day of symptoms, which could help define quarantine periods in infected individuals. In addition, it was found that the patient's Ct should not be considered an indicator of infectiousness, since it could not be correlated with the viral load disseminated. The results of this work are in line with proposed hypotheses of superspreaders, which can attribute part of the heterogeneity of the spread to the oversized emission of a small percentage of infected people.
Collapse
Affiliation(s)
- Marta Baselga
- Institute for Health Research Aragon (IIS Aragón), 50009 Zaragoza, Spain; (M.B.); (A.G.); (J.J.A.)
| | - Antonio Güemes
- Institute for Health Research Aragon (IIS Aragón), 50009 Zaragoza, Spain; (M.B.); (A.G.); (J.J.A.)
- Department of Surgery, University of Zaragoza, 50009 Zaragoza, Spain
| | - Juan J. Alba
- Institute for Health Research Aragon (IIS Aragón), 50009 Zaragoza, Spain; (M.B.); (A.G.); (J.J.A.)
- Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - Alberto J. Schuhmacher
- Institute for Health Research Aragon (IIS Aragón), 50009 Zaragoza, Spain; (M.B.); (A.G.); (J.J.A.)
- Fundación Agencia Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
203
|
Silva J, Patricio F, Patricio-Martínez A, Santos-López G, Cedillo L, Tizabi Y, Limón ID. Neuropathological Aspects of SARS-CoV-2 Infection: Significance for Both Alzheimer's and Parkinson's Disease. Front Neurosci 2022; 16:867825. [PMID: 35592266 PMCID: PMC9111171 DOI: 10.3389/fnins.2022.867825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Evidence suggests that SARS-CoV-2 entry into the central nervous system can result in neurological and/or neurodegenerative diseases. In this review, routes of SARS-Cov-2 entry into the brain via neuroinvasive pathways such as transcribrial, ocular surface or hematogenous system are discussed. It is argued that SARS-Cov-2-induced cytokine storm, neuroinflammation and oxidative stress increase the risk of developing neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Further studies on the effects of SARS-CoV-2 and its variants on protein aggregation, glia or microglia activation, and blood-brain barrier are warranted.
Collapse
Affiliation(s)
- Jaime Silva
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Felipe Patricio
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Gerardo Santos-López
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Atlixco, Mexico
| | - Lilia Cedillo
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
204
|
Deng Y, Li J, Sun C, Chi H, Luo D, Wang R, Qiu H, Zhang Y, Wu M, Zhang X, Huang X, Xie L, Qin C. Rational Development of a Polysaccharide-Protein-Conjugated Nanoparticle Vaccine Against SARS-CoV-2 Variants and Streptococcus pneumoniae. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200443. [PMID: 35332581 PMCID: PMC9073961 DOI: 10.1002/adma.202200443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/18/2022] [Indexed: 05/23/2023]
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 has led to millions of deaths worldwide. Streptococcus pneumoniae (S. pneumoniae) remains a major cause of mortality in underdeveloped countries. A vaccine that prevents both SARS-CoV-2 and S. pneumoniae infection represents a long-sought "magic bullet". Herein, a nanoparticle vaccine, termed SCTV01B, is rationally developed by using the capsular polysaccharide of S. pneumoniae serotype 14 (PPS14) as the backbone to conjugate with the recombinant receptor-binding domain (RBD) of the SARS-CoV-2 spike protein. The final formulation of conjugated nanoparticles in the network structure exhibits high thermal stability. Immunization with SCTV01B induces potent humoral and Type 1/Type 2 T helper cell (Th1/Th2) cellular immune responses in mice, rats, and rhesus macaques. In particular, SCTV01B-immunized serum not only broadly cross-neutralizes all SARS-CoV-2 variants of concern (VOCs), including the most recent Omicron variant, but also shows high opsonophagocytic activity (OPA) against S. pneumoniae serotype 14. Finally, SCTV01B vaccination confers protection against challenges with the SARS-CoV-2 mouse-adapted strain and the original strain in established murine models. Collectively, these promising preclinical results support further clinical evaluation of SCTV01B, highlighting the potency of polysaccharide-RBD-conjugated nanoparticle vaccine platforms for the development of vaccines for COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Yongqiang Deng
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Jing Li
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Chunyun Sun
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Hang Chi
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Dan Luo
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Rui Wang
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Hongying Qiu
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Yanjing Zhang
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Mei Wu
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Xiao Zhang
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Xun Huang
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
| | - Liangzhi Xie
- Beijing Protein and Antibody R&D Engineering CenterSinocelltech Ltd.Beijing100176P. R. China
| | - Chengfeng Qin
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAMMSBeijing100071P. R. China
- Research Unit of Discovery and Tracing of Natural Focus DiseasesChinese Academy of Medical SciencesBeijing100071P. R. China
| |
Collapse
|
205
|
Eicosanoid signalling blockade protects middle-aged mice from severe COVID-19. Nature 2022; 605:146-151. [PMID: 35314834 PMCID: PMC9783543 DOI: 10.1038/s41586-022-04630-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 03/11/2022] [Indexed: 12/27/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is especially severe in aged populations1. Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are highly effective, but vaccine efficacy is partly compromised by the emergence of SARS-CoV-2 variants with enhanced transmissibility2. The emergence of these variants emphasizes the need for further development of anti-SARS-CoV-2 therapies, especially for aged populations. Here we describe the isolation of highly virulent mouse-adapted viruses and use them to test a new therapeutic drug in infected aged animals. Many of the alterations observed in SARS-CoV-2 during mouse adaptation (positions 417, 484, 493, 498 and 501 of the spike protein) also arise in humans in variants of concern2. Their appearance during mouse adaptation indicates that immune pressure is not required for selection. For murine SARS, for which severity is also age dependent, elevated levels of an eicosanoid (prostaglandin D2 (PGD2)) and a phospholipase (phospholipase A2 group 2D (PLA2G2D)) contributed to poor outcomes in aged mice3,4. mRNA expression of PLA2G2D and prostaglandin D2 receptor (PTGDR), and production of PGD2 also increase with ageing and after SARS-CoV-2 infection in dendritic cells derived from human peripheral blood mononuclear cells. Using our mouse-adapted SARS-CoV-2, we show that middle-aged mice lacking expression of PTGDR or PLA2G2D are protected from severe disease. Furthermore, treatment with a PTGDR antagonist, asapiprant, protected aged mice from lethal infection. PTGDR antagonism is one of the first interventions in SARS-CoV-2-infected animals that specifically protects aged animals, suggesting that the PLA2G2D-PGD2/PTGDR pathway is a useful target for therapeutic interventions.
Collapse
|
206
|
Hong W, Yang J, Zou J, Bi Z, He C, Lei H, He X, Li X, Alu A, Ren W, Wang Z, Jiang X, Zhong K, Jia G, Yang Y, Yu W, Huang Q, Yang M, Zhou Y, Zhao Y, Kuang D, Wang J, Wang H, Chen S, Luo M, Zhang Z, Lu T, Chen L, Que H, He Z, Sun Q, Wang W, Shen G, Lu G, Zhao Z, Yang L, Yang J, Wang Z, Li J, Song X, Dai L, Chen C, Geng J, Gou M, Chen L, Dong H, Peng Y, Huang C, Qian Z, Cheng W, Fan C, Wei Y, Su Z, Tong A, Lu S, Peng X, Wei X. Histones released by NETosis enhance the infectivity of SARS-CoV-2 by bridging the spike protein subunit 2 and sialic acid on host cells. Cell Mol Immunol 2022; 19:577-587. [PMID: 35273357 PMCID: PMC8907557 DOI: 10.1038/s41423-022-00845-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023] Open
Abstract
Neutrophil extracellular traps (NETs) can capture and kill viruses, such as influenza viruses, human immunodeficiency virus (HIV), and respiratory syncytial virus (RSV), thus contributing to host defense. Contrary to our expectation, we show here that the histones released by NETosis enhance the infectivity of SARS-CoV-2, as found by using live SARS-CoV-2 and two pseudovirus systems as well as a mouse model. The histone H3 or H4 selectively binds to subunit 2 of the spike (S) protein, as shown by a biochemical binding assay, surface plasmon resonance and binding energy calculation as well as the construction of a mutant S protein by replacing four acidic amino acids. Sialic acid on the host cell surface is the key molecule to which histones bridge subunit 2 of the S protein. Moreover, histones enhance cell-cell fusion. Finally, treatment with an inhibitor of NETosis, histone H3 or H4, or sialic acid notably affected the levels of sgRNA copies and the number of apoptotic cells in a mouse model. These findings suggest that SARS-CoV-2 could hijack histones from neutrophil NETosis to promote its host cell attachment and entry process and may be important in exploring pathogenesis and possible strategies to develop new effective therapies for COVID-19.
Collapse
Affiliation(s)
- Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Jun Zou
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Xue Li
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Wenyan Ren
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Zeng Wang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Xiaohua Jiang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Kunhong Zhong
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Guowen Jia
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Yun Yang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Wenhai Yu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Qing Huang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Mengli Yang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Yanan Zhou
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Yuan Zhao
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Dexuan Kuang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Junbin Wang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Haixuan Wang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Ziqi Zhang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Tianqi Lu
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Zhiyao He
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Qiu Sun
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Wei Wang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Guobo Shen
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Zhiwei Zhao
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Li Yang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Jinliang Yang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Zhenling Wang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Jiong Li
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Xiangrong Song
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Lunzhi Dai
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Chong Chen
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Jia Geng
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Maling Gou
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Lu Chen
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Yong Peng
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Canhua Huang
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Wei Cheng
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, 102629, Beijing, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China
| | - Zhaoming Su
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Aiping Tong
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China.
| | - Shuaiyao Lu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.
| | - Xiaozhong Peng
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targeting, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, China.
- Westvac Biopharm Co., Ltd. No. 618, Fenghuang Road, Shuangliu District, Chengdu, Sichuan, China.
| |
Collapse
|
207
|
Treatment of SARS-CoV-2-induced pneumonia with NAD + and NMN in two mouse models. Cell Discov 2022; 8:38. [PMID: 35487885 PMCID: PMC9053567 DOI: 10.1038/s41421-022-00409-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/02/2022] [Indexed: 12/14/2022] Open
Abstract
The global COVID-19 epidemic has spread rapidly around the world and caused the death of more than 5 million people. It is urgent to develop effective strategies to treat COVID-19 patients. Here, we revealed that SARS-CoV-2 infection resulted in the dysregulation of genes associated with NAD+ metabolism, immune response, and cell death in mice, similar to that in COVID-19 patients. We therefore investigated the effect of treatment with NAD+ and its intermediate (NMN) and found that the pneumonia phenotypes, including excessive inflammatory cell infiltration, hemolysis, and embolization in SARS-CoV-2-infected lungs were significantly rescued. Cell death was suppressed substantially by NAD+ and NMN supplementation. More strikingly, NMN supplementation can protect 30% of aged mice infected with the lethal mouse-adapted SARS-CoV-2 from death. Mechanically, we found that NAD+ or NMN supplementation partially rescued the disturbed gene expression and metabolism caused by SARS-CoV-2 infection. Thus, our in vivo mouse study supports trials for treating COVID-19 patients by targeting the NAD+ pathway.
Collapse
|
208
|
Barman TK, Singh AK, Bonin JL, Nafiz TN, Salmon SL, Metzger DW. Lethal synergy between SARS-CoV-2 and Streptococcus pneumoniae in hACE2 mice and protective efficacy of vaccination. JCI Insight 2022; 7:159422. [PMID: 35482422 DOI: 10.1172/jci.insight.159422] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Secondary infections are frequent complications of viral respiratory infections but the potential consequence of SARS-CoV-2 co-infection with common pulmonary pathogens is poorly understood. We report that co-infection of human ACE2 transgenic mice with sublethal doses of SARS-CoV-2 and Streptococcus pneumoniae results in synergistic lung inflammation and lethality. Mortality was observed regardless of whether SARS-CoV-2 challenge occurred before or after establishment of sublethal pneumococcal infection. Increased bacterial levels following co-infection were associated with alveolar macrophage depletion and treatment with murine GM-CSF reduced lung bacteria numbers and pathology, and partially protected from death. However, therapeutic targeting of interferons, an approach that is effective against influenza co-infections, failed to increase survival. Combined vaccination against both SARS-CoV-2 and pneumococci resulted in 100% protection against subsequent co-infection. The results indicate that when seasonal respiratory infections return to pre-pandemic levels, they could lead to an increased incidence of lethal COVID-19 superinfections, especially among the unvaccinated population. .
Collapse
Affiliation(s)
- Tarani Kanta Barman
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Amit K Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Jesse L Bonin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Tanvir N Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Sharon L Salmon
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Dennis W Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| |
Collapse
|
209
|
Stegmann KM, Dickmanns A, Heinen N, Blaurock C, Karrasch T, Breithaupt A, Klopfleisch R, Uhlig N, Eberlein V, Issmail L, Herrmann ST, Schreieck A, Peelen E, Kohlhof H, Sadeghi B, Riek A, Speakman JR, Groß U, Görlich D, Vitt D, Müller T, Grunwald T, Pfaender S, Balkema-Buschmann A, Dobbelstein M. Inhibitors of dihydroorotate dehydrogenase cooperate with Molnupiravir and N4-hydroxycytidine to suppress SARS-CoV-2 replication. iScience 2022; 25:104293. [PMID: 35492218 PMCID: PMC9035612 DOI: 10.1016/j.isci.2022.104293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
The nucleoside analog N4-hydroxycytidine (NHC) is the active metabolite of the prodrug molnupiravir, which has been approved for the treatment of COVID-19. SARS-CoV-2 incorporates NHC into its RNA, resulting in defective virus genomes. Likewise, inhibitors of dihydroorotate dehydrogenase (DHODH) reduce virus yield upon infection, by suppressing the cellular synthesis of pyrimidines. Here, we show that NHC and DHODH inhibitors strongly synergize in the inhibition of SARS-CoV-2 replication in vitro. We propose that the lack of available pyrimidine nucleotides upon DHODH inhibition increases the incorporation of NHC into nascent viral RNA. This concept is supported by the rescue of virus replication upon addition of pyrimidine nucleosides to the media. DHODH inhibitors increased the antiviral efficiency of molnupiravir not only in organoids of human lung, but also in Syrian Gold hamsters and in K18-hACE2 mice. Combining molnupiravir with DHODH inhibitors may thus improve available therapy options for COVID-19. Molnupiravir and DHODH inhibitors are approved drugs, facilitating clinical testing The combination may allow lower drug doses to decrease possible toxic effects Inhibitors of nucleotide biosynthesis may boost antiviral nucleoside analogs
Collapse
Affiliation(s)
- Kim M Stegmann
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Antje Dickmanns
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Natalie Heinen
- Department of Molecular and Medical Virology, Ruhr University Bochum, Germany
| | - Claudia Blaurock
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald - Insel Riems, Germany
| | - Tim Karrasch
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Angele Breithaupt
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald - Insel Riems, Germany
| | | | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Simon T Herrmann
- Department of Molecular Biochemistry, Ruhr University Bochum, Germany
| | | | | | | | - Balal Sadeghi
- Friedrich-Loeffler-Institut, Institute of Novel and Emerging Infectious Diseases, Greifswald - Insel Riems, Germany
| | - Alexander Riek
- Friedrich-Loeffler-Institut, Institute of Animal Welfare and Animal Husbandry, Celle, Germany
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, UK
| | - Uwe Groß
- Institute of Medical Microbiology and Virology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Germany
| | - Dirk Görlich
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | | | - Thorsten Müller
- Department of Molecular Biochemistry, Ruhr University Bochum, Germany.,Institute of Psychiatric Phenomics and Genomics (IPPG), Organoid laboratory, University Hospital, LMU Munich, Germany
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Germany
| | - Anne Balkema-Buschmann
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald - Insel Riems, Germany
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| |
Collapse
|
210
|
MacDougall M, El-Hajj Sleiman J, Beauchemin P, Rangachari M. SARS-CoV-2 and Multiple Sclerosis: Potential for Disease Exacerbation. Front Immunol 2022; 13:871276. [PMID: 35572514 PMCID: PMC9102605 DOI: 10.3389/fimmu.2022.871276] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
While the respiratory tract is the primary route of entry for SARS-CoV-2, evidence shows that the virus also impacts the central nervous system. Intriguingly, case reports have documented SARS-CoV-2 patients presenting with demyelinating lesions in the brain, spinal cord, and optic nerve, suggesting possible implications in neuroimmune disorders such as multiple sclerosis (MS) and other related neuroimmune disorders. However, the cellular mechanisms underpinning these observations remain poorly defined. The goal of this paper was to review the literature to date regarding possible links between SARS-CoV-2 infection and neuroimmune demyelinating diseases such as MS and its related disorders, with the aim of positing a hypothesis for disease exacerbation. The literature suggests that SARS-CoV, SARS-CoV-2, and orthologous murine coronaviruses invade the CNS via the olfactory bulb, spreading to connected structures via retrograde transport. We hypothesize that a glial inflammatory response may contribute to damaged oligodendrocytes and blood brain barrier (BBB) breakdown, allowing a second route for CNS invasion and lymphocyte infiltration. Potential for molecular mimicry and the stimulation of autoreactive T cells against myelin is also described. It is imperative that further studies on SARS-CoV-2 neuroinvasion address the adverse effects of the virus on myelin and exacerbation of MS symptoms, as nearly 3 million people suffer from MS worldwide.
Collapse
Affiliation(s)
- Madison MacDougall
- Department of Biological Sciences, Salisbury University, Salisbury, MD, United States
- Department of Psychology, Salisbury University, Salisbury, MD, United States
| | - Jad El-Hajj Sleiman
- Division of Neurology, Department of Medicine, CHU de Québec – Université Laval, Quebec City, QC, Canada
| | - Philippe Beauchemin
- Division of Neurology, Department of Medicine, CHU de Québec – Université Laval, Quebec City, QC, Canada
| | - Manu Rangachari
- Axe Neurosciences, Centre de Recherche du CHU de Québec – Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
211
|
Platelet activation by SARS-CoV-2 implicates the release of active tissue factor by infected cells. Blood Adv 2022; 6:3593-3605. [PMID: 35443030 PMCID: PMC9023084 DOI: 10.1182/bloodadvances.2022007444] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 11/20/2022] Open
Abstract
Platelets are hyperactivated in coronavirus disease 2019 (COVID-19). However, the mechanisms promoting platelet activation by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are not well understood. This may be due to inherent challenges in discriminating the contribution of viral vs host components produced by infected cells. This is particularly true for enveloped viruses and extracellular vesicles (EVs), as they are concomitantly released during infection and share biophysical properties. To study this, we evaluated whether SARS-CoV-2 itself or components derived from SARS-CoV-2-infected human lung epithelial cells could activate isolated platelets from healthy donors. Activation was measured by the surface expression of P-selectin and the activated conformation of integrin αIIbβ3, degranulation, aggregation under flow conditions, and the release of EVs. We find that neither SARS-CoV-2 nor purified spike activates platelets. In contrast, tissue factor (TF) produced by infected cells was highly potent at activating platelets. This required trace amounts of plasma containing the coagulation factors FX, FII, and FVII. Robust platelet activation involved thrombin and the activation of protease-activated receptor (PAR)-1 and -4 expressed by platelets. Virions and EVs were identified by electron microscopy. Through size-exclusion chromatography, TF activity was found to be associated with a virus or EVs, which were indistinguishable. Increased TF messenger RNA (mRNA) expression and activity were also found in lungs in a murine model of COVID-19 and plasma of severe COVID-19 patients, respectively. In summary, TF activity from SARS-CoV-2–infected cells activates thrombin, which signals to PARs on platelets. Blockade of molecules in this pathway may interfere with platelet activation and the coagulation characteristic of COVID-19.
Collapse
|
212
|
Chernov AS, Minakov AA, Kazakov VA, Rodionov MV, Rybalkin IN, Vlasik TN, Yashin DV, Saschenko LP, Kudriaeva AA, Belogurov AA, Smirnov IV, Loginova SY, Schukina VN, Savenko SV, Borisevich SV, Zykov KA, Gabibov AG, Telegin GB. A new mouse unilateral model of diffuse alveolar damage of the lung. Inflamm Res 2022; 71:627-639. [PMID: 35434745 PMCID: PMC9013507 DOI: 10.1007/s00011-022-01568-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Objective and design The existing biological models of diffuse alveolar damage (DAD) in mice have many shortcomings. To offset these shortcomings, we have proposed a simple, nonsurgical, and reproducible method of unilateral total damage of the left lung in ICR mice. This model is based on the intrabronchial administration of a mixture of bacterial lipopolysaccharide (LPS) from the cell wall of S. enterica and α-galactosylceramide (inducing substances) to the left lung. Methods Using computer tomography of the lungs with endobronchial administration of contrast material, we have been able to perform an operative intravital verification of the targeted delivery of the inducer. The model presented is characterized by more serious and homogeneous damage of the affected lung compared to the existing models of focal pneumonia; at the same time, our model is characterized by longer animal survival since the right lung remains intact. Results The model is also characterized by diffuse alveolar damage of the left lung, animal survival of 100%, abrupt increases in plasma levels of TNFa, INFg, and IL-6, and significant myocardial overload in the right heart. It can be used to assess the efficacy of innovative drugs for the treatment of DAD and ARDS as the clinical manifestations that are developed in patients infected with SARS-CoV-2. Morphological patterns of lungs in the noninfectious (“sterile”) model of DAD induced by LPS simultaneously with α-galactosylceramide (presented here) and in the infectious model of DAD induced by SARS-CoV-2 have been compared. Conclusion The DAD model we have proposed can be widely used for studying the efficacy of candidate molecules for the treatment of infectious respiratory diseases, such as viral pneumonias of different etiology, including SARS-CoV-2.
Collapse
|
213
|
Muhl L, He L, Sun Y, Andaloussi Mäe M, Pietilä R, Liu J, Genové G, Zhang L, Xie Y, Leptidis S, Mocci G, Stritt S, Osman A, Anisimov A, Hemanthakumar KA, Räsänen M, Hansson EM, Björkegren J, Vanlandewijck M, Blomgren K, Mäkinen T, Peng XR, Hu Y, Ernfors P, Arnold TD, Alitalo K, Lendahl U, Betsholtz C. The SARS-CoV-2 receptor ACE2 is expressed in mouse pericytes but not endothelial cells: Implications for COVID-19 vascular research. Stem Cell Reports 2022; 17:1089-1104. [PMID: 35452595 PMCID: PMC9022216 DOI: 10.1016/j.stemcr.2022.03.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
Humanized mouse models and mouse-adapted SARS-CoV-2 virus are increasingly used to study COVID-19 pathogenesis, so it is important to learn where the SARS-CoV-2 receptor ACE2 is expressed. Here we mapped ACE2 expression during mouse postnatal development and in adulthood. Pericytes in the CNS, heart, and pancreas express ACE2 strongly, as do perineurial and adrenal fibroblasts, whereas endothelial cells do not at any location analyzed. In a number of other organs, pericytes do not express ACE2, including in the lung where ACE2 instead is expressed in bronchial epithelium and alveolar type II cells. The onset of ACE2 expression is organ specific: in bronchial epithelium already at birth, in brain pericytes before, and in heart pericytes after postnatal day 10.5. Establishing the vascular localization of ACE2 expression is central to correctly interpret data from modeling COVID-19 in the mouse and may shed light on the cause of vascular COVID-19 complications. Detailed Ace2/ACE2 expression patterns are reported for multiple mouse organs Vascular Ace2/ACE2 expression occurs in pericytes but not endothelial cells Ace2/ACE2 expression is organotypic and developmentally regulated Ace2/ACE2 expression in pericytes may suggest their involvement in COVID-19
Collapse
Affiliation(s)
- Lars Muhl
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Liqun He
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Ying Sun
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Riikka Pietilä
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Jianping Liu
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Guillem Genové
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Lei Zhang
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an China
| | - Yuan Xie
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an China
| | - Stefanos Leptidis
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Giuseppe Mocci
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Simon Stritt
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ahmed Osman
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Markus Räsänen
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Emil M Hansson
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Johan Björkegren
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden; Institute of Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Vanlandewijck
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden; Department of Pediatric Oncology, Karolinska University Hospital, Stockholm Sweden
| | - Taija Mäkinen
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Xiao-Rong Peng
- Cardiovascular, Renal and Metabolism, AstraZeneca BioPharmaceutical R&D, Gothenburg, Sweden
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Thomas D Arnold
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden.
| | - Christer Betsholtz
- Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
214
|
Affiliation(s)
- Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, and Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China. .,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China. .,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, and Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.,Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Academician Workstation of Hainan Province, Hainan Medical University, Haikou, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, and Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China. .,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China. .,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China. .,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China. .,Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China. .,Academician Workstation of Hainan Province, Hainan Medical University, Haikou, China.
| |
Collapse
|
215
|
A potent human monoclonal antibody with pan-neutralizing activities directly dislocates S trimer of SARS-CoV-2 through binding both up and down forms of RBD. Signal Transduct Target Ther 2022; 7:114. [PMID: 35383141 PMCID: PMC8980211 DOI: 10.1038/s41392-022-00954-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 11/09/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of novel coronavirus disease (COVID-19). The neutralizing monoclonal antibodies (mAbs) targeting the receptor-binding domain (RBD) of SARS-CoV-2 are among the most promising strategies to prevent and treat COVID-19. However, SARS-CoV-2 variants of concern (VOCs) profoundly reduced the efficacies of most of mAbs and vaccines approved for clinical use. Herein, we demonstrated mAb 35B5 efficiently neutralizes both wild-type (WT) SARS-CoV-2 and VOCs, including B.1.617.2 (delta) variant, in vitro and in vivo. Cryo-electron microscopy (cryo-EM) revealed that 35B5 neutralizes SARS-CoV-2 by targeting a unique epitope that avoids the prevailing mutation sites on RBD identified in circulating VOCs, providing the molecular basis for its pan-neutralizing efficacy. The 35B5-binding epitope could also be exploited for the rational design of a universal SARS-CoV-2 vaccine.
Collapse
|
216
|
Two Different Therapeutic Approaches for SARS-CoV-2 in hiPSCs-Derived Lung Organoids. Cells 2022; 11:cells11071235. [PMID: 35406799 PMCID: PMC8997767 DOI: 10.3390/cells11071235] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/21/2022] [Accepted: 04/02/2022] [Indexed: 12/14/2022] Open
Abstract
The global health emergency for SARS-CoV-2 (COVID-19) created an urgent need to develop new treatments and therapeutic drugs. In this study, we tested, for the first time on human cells, a new tetravalent neutralizing antibody (15033-7) targeting Spike protein and a synthetic peptide homologous to dipeptidyl peptidase-4 (DPP4) receptor on host cells. Both could represent powerful immunotherapeutic candidates for COVID-19 treatment. The infection begins in the proximal airways, namely the alveolar type 2 (AT2) cells of the distal lung, which express both ACE2 and DPP4 receptors. Thus, to evaluate the efficacy of both approaches, we developed three-dimensional (3D) complex lung organoid structures (hLORGs) derived from human-induced pluripotent stem cells (iPSCs) and resembling the in vivo organ. Afterward, hLORGs were infected by different SARS-CoV-2 S pseudovirus variants and treated by the Ab15033-7 or DPP4 peptide. Using both approaches, we observed a significant reduction of viral entry and a modulation of the expression of genes implicated in innate immunity and inflammatory response. These data demonstrate the efficacy of such approaches in strongly reducing the infection efficiency in vitro and, importantly, provide proof-of-principle evidence that hiPSC-derived hLORGs represent an ideal in vitro system for testing both therapeutic and preventive modalities against COVID-19.
Collapse
|
217
|
COVID-19: A Veterinary and One Health Perspective. J Indian Inst Sci 2022; 102:689-709. [PMID: 35968231 PMCID: PMC9364302 DOI: 10.1007/s41745-022-00318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 06/21/2022] [Indexed: 10/30/2022]
Abstract
Interface with animals has been responsible for the occurrence of a major proportion of human diseases for the past several decades. Recent outbreaks of respiratory, haemorrhagic, encephalitic, arthropod-borne and other viral diseases have underlined the role of animals in the transmission of pathogens to humans. The on-going coronavirus disease-2019 (COVID-19) pandemic is one among them and is thought to have originated from bats and jumped to humans through an intermediate animal host. Indeed, the aetiology, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can infect and cause disease in cats, ferrets and minks, as well as be transmitted from one animal to another. The seriousness of the pandemic along with the zoonotic origin of the virus has been a red alert on the critical need for collaboration and cooperation among human and animal health professionals, as well as stakeholders from various other disciplines that study planetary health parameters and the well-being of the biosphere. It is therefore imminent that One Health principles are applied across the board for human infectious diseases so that we can be better prepared for future zoonotic disease outbreaks and pandemics.
Collapse
|
218
|
Al-Ani B, ShamsEldeen AM, Kamar SS, Haidara MA, Al-Hashem F, Alshahrani MY, Al-Hakami AM, Kader DHA, Maarouf A. Lipopolysaccharide induces acute lung injury and alveolar haemorrhage in association with the cytokine storm, coagulopathy and AT1R/JAK/STAT augmentation in a rat model that mimics moderate and severe Covid-19 pathology. Clin Exp Pharmacol Physiol 2022; 49:483-491. [PMID: 35066912 DOI: 10.1111/1440-1681.13620] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023]
Abstract
Progress in the study of Covid-19 disease in rodents has been hampered by the lack of angiotensin-converting enzyme 2 (ACE2; virus entry route to the target cell) affinities for the virus spike proteins across species. Therefore, we sought to determine whether a modified protocol of lipopolysaccharide (LPS)-induced acute respiratory distress syndrome in rats can mimic both cell signalling pathways as well as severe disease phenotypes of Covid-19 disease. Rats were injected via intratracheal (IT) instillation with either 15 mg/kg of LPS (model group) or saline (control group) before being killed after 3 days. A severe acute respiratory syndrome (SARS)-like effect was observed in the model group as demonstrated by the development of a "cytokine storm" (>2.7 fold increase in blood levels of IL-6, IL-17A, GM-CSF, and TNF-α), high blood ferritin, demonstrable coagulopathy, including elevated D-dimer (approximately 10-fold increase), PAI-1, PT, and APTT (p < 0.0001). In addition, LPS increased the expression of lung angiotensin II type I receptor (AT1R)-JAK-STAT axis (>4 fold increase). Chest imaging revealed bilateral small patchy opacities of the lungs. Severe lung injury was noted by the presence of both, alveolar collapse and haemorrhage, desquamation of epithelial cells in the airway lumen, infiltration of inflammatory cells (CD45+ leukocytes), widespread thickening of the interalveolar septa, and ultrastructural alterations similar to Covid-19. Thus, these findings demonstrate that IT injection of 15 mg/kg LPS into rats, induced an AT1R/JAK/STAT-mediated cytokine storm with resultant pneumonia and coagulopathy that was commensurate with moderate and severe Covid-19 disease noted in humans.
Collapse
Affiliation(s)
- Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asmaa M ShamsEldeen
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samaa S Kamar
- Department of Medical Histology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A Haidara
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Fahaid Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ahmed M Al-Hakami
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Dina H Abdel Kader
- Department of Medical Histology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amro Maarouf
- Department of Clinical Biochemistry, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
219
|
Bain CC, Lucas CD, Rossi AG. Pulmonary macrophages and SARS-Cov2 infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 367:1-28. [PMID: 35461655 PMCID: PMC8968207 DOI: 10.1016/bs.ircmb.2022.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the largest global pandemic in living memory, with between 4.5 and 15M deaths globally from coronavirus disease 2019 (COVID-19). This has led to an unparalleled global, collaborative effort to understand the pathogenesis of this devastating disease using state-of-the-art technologies. A consistent feature of severe COVID-19 is dysregulation of pulmonary macrophages, cells that under normal physiological conditions play vital roles in maintaining lung homeostasis and immunity. In this article, we will discuss a selection of the pivotal findings examining the role of monocytes and macrophages in SARS-CoV-2 infection and place this in context of recent advances made in understanding the fundamental immunobiology of these cells to try to understand how key homeostatic cells come to be a central pathogenic component of severe COVID-19 and key cells to target for therapeutic gain.
Collapse
Affiliation(s)
- Calum C Bain
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, United Kingdom; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom.
| | - Christopher D Lucas
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, United Kingdom; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom.
| | - Adriano G Rossi
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, United Kingdom; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom.
| |
Collapse
|
220
|
Ziuzia-Januszewska L, Januszewski M. Pathogenesis of Olfactory Disorders in COVID-19. Brain Sci 2022; 12:brainsci12040449. [PMID: 35447981 PMCID: PMC9029941 DOI: 10.3390/brainsci12040449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Since the outbreak of the SARS-CoV-2 pandemic, olfactory disorders have been reported as a frequent symptom of COVID-19; however, its pathogenesis is still debated. The aim of this review is to summarize the current understanding of the pathogenesis of smell impairment in the course of COVID-19 and to highlight potential avenues for future research on this issue. Several theories have been proposed to explain the pathogenesis of COVID-19-related anosmia, including nasal obstruction and rhinorrhea, oedema of the olfactory cleft mucosa, olfactory epithelial damage either within the olfactory receptor cells or the supporting non-neural cells (either direct or immune-mediated), damage to the olfactory bulb, and impairment of the central olfactory pathways. Although the pathogenesis of COVID-19-related anosmia is still not fully elucidated, it appears to be mainly due to sensorineural damage, with infection of the olfactory epithelium support cells via the ACE1 receptor and disruption of the OE caused by immense inflammatory reaction, and possibly with direct olfactory sensory neurons infection mediated by the NRP-1 receptor. Involvement of the higher olfactory pathways and a conductive component of olfactory disorders, as well as genetic factors, may also be considered.
Collapse
Affiliation(s)
- Laura Ziuzia-Januszewska
- Department of Otolaryngology, Central Clinical Hospital, Ministry of Interior and Administration, 02-507 Warsaw, Poland
- Correspondence: or ; Tel.: +48-477221182
| | - Marcin Januszewski
- Department of Obstetrics and Gynecology, Central Clinical Hospital, Ministry of Interior and Administration, 02-507 Warsaw, Poland;
| |
Collapse
|
221
|
Wang P, Zhou R, Zhou R, Li W, Weerasinghe J, Chen S, Rehm BHA, Zhao L, Frentiu FD, Zhang Z, Yan K, Lor M, Suhrbier A, Richard DJ, Thompson EW, Ostrikov KK, Dai X. Cold atmospheric plasma for preventing infection of viruses that use ACE2 for entry. Theranostics 2022; 12:2811-2832. [PMID: 35401827 PMCID: PMC8965494 DOI: 10.7150/thno.70098] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/17/2022] [Indexed: 11/05/2022] Open
Abstract
Rational: The mutating SARS-CoV-2 potentially impairs the efficacy of current vaccines or antibody-based treatments. Broad-spectrum and rapid anti-virus methods feasible for regular epidemic prevention against COVID-19 or alike are urgently called for. Methods: Using SARS-CoV-2 virus and bioengineered pseudoviruses carrying ACE2-binding spike protein domains, we examined the efficacy of cold atmospheric plasma (CAP) on virus entry prevention. Results: We found that CAP could effectively inhibit the entry of virus into cells. Direct CAP or CAP-activated medium (PAM) triggered rapid internalization and nuclear translocation of the virus receptor, ACE2, which began to return after 5 hours and was fully recovered by 12 hours. This was seen in vitro with both VERO-E6 cells and human mammary epithelial MCF10A cells, and in vivo. Hydroxyl radical (·OH) and species derived from its interactions with other species were found to be the most effective CAP components for triggering ACE2 nucleus translocation. The ERα/STAT3(Tyr705) and EGFR(Tyr1068/1086)/STAT3(Tyr705) axes were found to interact and collectively mediate the effects on ACE2 localization and expression. Conclusions: Our data support the use of PAM in helping control SARS-CoV-2 if developed into products for nose/mouth spray; an approach extendable to other viruses utilizing ACE2 for host entry.
Collapse
Affiliation(s)
- Peiyu Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
- Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Renwu Zhou
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
- State Key Laboratory of Electrical Insulation and Power Equipment, Center for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Rusen Zhou
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Wenshao Li
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Janith Weerasinghe
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Shuxiong Chen
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Liqian Zhao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Francesca D. Frentiu
- School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4059, Australia
| | - Zhifa Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Kexin Yan
- QIMR Berghofer Medical Research Institute, Herston QLD 4006, Australia
| | - Mary Lor
- QIMR Berghofer Medical Research Institute, Herston QLD 4006, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Herston QLD 4006, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, Queensland, Australia
| | - Derek J. Richard
- School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
- Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Erik W. Thompson
- School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
- Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- CAPsoul Biotechnology Company, Ltd, Beijing, China
| |
Collapse
|
222
|
Qin S, Li R, Zheng Z, Zeng X, Wang Y, Wang X. Review of selected animal models for respiratory coronavirus infection and its application in drug research. J Med Virol 2022; 94:3032-3042. [PMID: 35285034 PMCID: PMC9088459 DOI: 10.1002/jmv.27718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Numerous viral pneumonia cases have been reported in Wuhan, Hubei in December 2019. The pathogen has been identified as a novel coronavirus, which was named severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). The biological characteristics and pathogenesis mechanism of SARS‐CoV‐2 are unclear and under progress. At present, no specific preventive and therapeutic drugs are available. Animal models can reproduce the viral replication cycle and the significant functions of respiratory coronavirus infection and are urgently needed to evaluate the efficacy of drugs and vaccines, the transmission route of respiratory coronavirus, clinical features, and so on. We reviewed the current animal models of respiratory coronavirus (SARS‐CoV, MERS‐CoV, and SARS‐CoV‐2) infection and made a comparative analysis of the route of inoculation, virus replication, clinical signs, histopathology, application, advantages, and disadvantages. Animal models of respiratory coronavirus include susceptible animal models, genetically modified models, and various animal models of infected virus adaptation strains, such as nonhuman primates, mice, hamsters, ferrets, New Zealand rabbits, cats, and other animal models, all of which have distinct advantages and limitations. This review will provide relevant information and important insights for disease management and control. Animal models for coronavirus infection.
Collapse
Affiliation(s)
- Shengle Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, GuangdongChina
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, GuangdongChina
| | | | - Xuxin Zeng
- School of MedicineFoshan UniversityFoshanChina
| | - Yutao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, GuangdongChina
| | - Xinhua Wang
- School of MedicineFoshan UniversityFoshanChina
| |
Collapse
|
223
|
Carossino M, Kenney D, O’Connell AK, Montanaro P, Tseng AE, Gertje HP, Grosz KA, Ericsson M, Huber BR, Kurnick SA, Subramaniam S, Kirkland TA, Walker JR, Francis KP, Klose AD, Paragas N, Bosmann M, Saeed M, Balasuriya UBR, Douam F, Crossland NA. Fatal Neurodissemination and SARS-CoV-2 Tropism in K18-hACE2 Mice Is Only Partially Dependent on hACE2 Expression. Viruses 2022; 14:v14030535. [PMID: 35336942 PMCID: PMC8955233 DOI: 10.3390/v14030535] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/17/2022] [Accepted: 03/03/2022] [Indexed: 02/06/2023] Open
Abstract
Animal models recapitulating COVID-19 are critical to enhance our understanding of SARS-CoV-2 pathogenesis. Intranasally inoculated transgenic mice expressing human angiotensin-converting enzyme 2 under the cytokeratin 18 promoter (K18-hACE2) represent a lethal model of SARS-CoV-2 infection. We evaluated the clinical and virological dynamics of SARS-CoV-2 using two intranasal doses (104 and 106 PFUs), with a detailed spatiotemporal pathologic analysis of the 106 dose cohort. Despite generally mild-to-moderate pneumonia, clinical decline resulting in euthanasia or death was commonly associated with hypothermia and viral neurodissemination independent of inoculation dose. Neuroinvasion was first observed at 4 days post-infection, initially restricted to the olfactory bulb suggesting axonal transport via the olfactory neuroepithelium as the earliest portal of entry. Absence of viremia suggests neuroinvasion occurs independently of transport across the blood-brain barrier. SARS-CoV-2 tropism was neither restricted to ACE2-expressing cells (e.g., AT1 pneumocytes), nor inclusive of some ACE2-positive cell lineages (e.g., bronchiolar epithelium and brain vasculature). Absence of detectable ACE2 protein expression in neurons but overexpression in neuroepithelium suggest this as the most likely portal of neuroinvasion, with subsequent ACE2 independent lethal neurodissemination. A paucity of epidemiological data and contradicting evidence for neuroinvasion and neurodissemination in humans call into question the translational relevance of this model.
Collapse
Affiliation(s)
- Mariano Carossino
- Louisiana Animal Disease Diagnostic Laboratory (LADDL), Louisiana State University, Baton Rouge, LA 61329, USA; (M.C.); (U.B.R.B.)
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 61329, USA
| | - Devin Kenney
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Aoife K. O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
| | - Paige Montanaro
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anna E. Tseng
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hans P. Gertje
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
| | - Kyle A. Grosz
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
| | - Maria Ericsson
- Electron Microscopy Core Facility, Harvard Medical School, Boston, MA 02115, USA;
| | - Bertrand R. Huber
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Susanna A. Kurnick
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
| | - Saravanan Subramaniam
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Thomas A. Kirkland
- Promega Biosciences, LLC, San Luis Obispo, CA 93401, USA; (T.A.K.); (J.R.W.)
| | - Joel R. Walker
- Promega Biosciences, LLC, San Luis Obispo, CA 93401, USA; (T.A.K.); (J.R.W.)
| | | | | | - Neal Paragas
- InVivo Analytics Inc., New York, NY 10023, USA; (A.D.K.); (N.P.)
- Department of Radiology Imaging Research Lab, University of Washington, Seattle, WA 98133, USA
| | - Markus Bosmann
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA;
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Biochemistry, Boston University, Boston, MA 02118, USA
| | - Udeni B. R. Balasuriya
- Louisiana Animal Disease Diagnostic Laboratory (LADDL), Louisiana State University, Baton Rouge, LA 61329, USA; (M.C.); (U.B.R.B.)
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 61329, USA
| | - Florian Douam
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: (F.D.); (N.A.C.); Tel.: +1-(617)-358-9174 (F.D.); +1-(617)-358-9285 (N.A.C.)
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA; (D.K.); (A.K.O.); (P.M.); (A.E.T.); (H.P.G.); (K.A.G.); (S.A.K.); (M.B.); (M.S.)
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: (F.D.); (N.A.C.); Tel.: +1-(617)-358-9174 (F.D.); +1-(617)-358-9285 (N.A.C.)
| |
Collapse
|
224
|
Jafari A, Danesh Pouya F, Niknam Z, Abdollahpour-Alitappeh M, Rezaei-Tavirani M, Rasmi Y. Current advances and challenges in COVID-19 vaccine development: from conventional vaccines to next-generation vaccine platforms. Mol Biol Rep 2022; 49:4943-4957. [PMID: 35235159 PMCID: PMC8890022 DOI: 10.1007/s11033-022-07132-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
The world is grappling with an unprecedented public health crisis COVID-19 pandemic caused by the novel coronavirus SARS-CoV-2. Due to the high transmission/mortality rates and socioeconomic impacts of the COVID-19, its control is crucial. In the absence of specific treatment, vaccines represent the most efficient way to control and stop the pandemic. Many companies around the world are currently making efforts to develop the vaccine to combat COVID-19. This review outlines key strategies for generating SARS-CoV-2 vaccine candidates, along with the mechanism of action, advantages, and potential limitations of each vaccine. The use of nanomaterials and nanotechnology for COVID-19 vaccines development will also be discussed.
Collapse
Affiliation(s)
- Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.,Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahima Danesh Pouya
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Niknam
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meghdad Abdollahpour-Alitappeh
- Cellular and Molecular Biology Research Center, Larestan University of Medical Sciences, Larestan, Iran.,Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
225
|
Liu J, Lu F, Chen Y, Plow E, Qin J. Integrin mediates cell entry of the SARS-CoV-2 virus independent of cellular receptor ACE2. J Biol Chem 2022; 298:101710. [PMID: 35150743 PMCID: PMC8828381 DOI: 10.1016/j.jbc.2022.101710] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It is broadly accepted that SARS-CoV-2 utilizes its spike protein to recognize the extracellular domain of angiotensin-converting enzyme 2 (ACE2) to enter cells for viral infection. However, other mechanisms of SARS-CoV-2 cell entry may occur. We show quantitatively that the SARS-CoV-2 spike protein also binds to the extracellular domain of broadly expressed integrin α5β1 with an affinity comparable to that of SARS-CoV-2 binding to ACE2. More importantly, we provide direct evidence that such binding promotes the internalization of SARS-CoV-2 into non-ACE2 cells in a manner critically dependent upon the activation of the integrin. Our data demonstrate an alternative pathway for the cell entry of SARS-CoV-2, suggesting that upon initial ACE2-mediated invasion of the virus in the respiratory system, which is known to trigger an immune response and secretion of cytokines to activate integrin, the integrin-mediated cell invasion of SARS-CoV-2 into the respiratory system and other organs becomes effective, thereby promoting further infection and progression of COVID-19.
Collapse
Affiliation(s)
- Jiamnin Liu
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fan Lu
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yinghua Chen
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Edward Plow
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jun Qin
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
226
|
Aghamirza Moghim Aliabadi H, Eivazzadeh‐Keihan R, Beig Parikhani A, Fattahi Mehraban S, Maleki A, Fereshteh S, Bazaz M, Zolriasatein A, Bozorgnia B, Rahmati S, Saberi F, Yousefi Najafabadi Z, Damough S, Mohseni S, Salehzadeh H, Khakyzadeh V, Madanchi H, Kardar GA, Zarrintaj P, Saeb MR, Mozafari M. COVID-19: A systematic review and update on prevention, diagnosis, and treatment. MedComm (Beijing) 2022; 3:e115. [PMID: 35281790 PMCID: PMC8906461 DOI: 10.1002/mco2.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 01/09/2023] Open
Abstract
Since the rapid onset of the COVID-19 or SARS-CoV-2 pandemic in the world in 2019, extensive studies have been conducted to unveil the behavior and emission pattern of the virus in order to determine the best ways to diagnosis of virus and thereof formulate effective drugs or vaccines to combat the disease. The emergence of novel diagnostic and therapeutic techniques considering the multiplicity of reports from one side and contradictions in assessments from the other side necessitates instantaneous updates on the progress of clinical investigations. There is also growing public anxiety from time to time mutation of COVID-19, as reflected in considerable mortality and transmission, respectively, from delta and Omicron variants. We comprehensively review and summarize different aspects of prevention, diagnosis, and treatment of COVID-19. First, biological characteristics of COVID-19 were explained from diagnosis standpoint. Thereafter, the preclinical animal models of COVID-19 were discussed to frame the symptoms and clinical effects of COVID-19 from patient to patient with treatment strategies and in-silico/computational biology. Finally, the opportunities and challenges of nanoscience/nanotechnology in identification, diagnosis, and treatment of COVID-19 were discussed. This review covers almost all SARS-CoV-2-related topics extensively to deepen the understanding of the latest achievements (last updated on January 11, 2022).
Collapse
Affiliation(s)
- Hooman Aghamirza Moghim Aliabadi
- Protein Chemistry LaboratoryDepartment of Medical BiotechnologyBiotechnology Research CenterPasteur Institute of IranTehranIran
- Advance Chemical Studies LaboratoryFaculty of ChemistryK. N. Toosi UniversityTehranIran
| | | | - Arezoo Beig Parikhani
- Department of Medical BiotechnologyBiotechnology Research CenterPasteur InstituteTehranIran
| | | | - Ali Maleki
- Department of ChemistryIran University of Science and TechnologyTehranIran
| | | | - Masoume Bazaz
- Department of Medical BiotechnologyBiotechnology Research CenterPasteur InstituteTehranIran
| | | | | | - Saman Rahmati
- Department of Medical BiotechnologyBiotechnology Research CenterPasteur InstituteTehranIran
| | - Fatemeh Saberi
- Department of Medical BiotechnologySchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zeinab Yousefi Najafabadi
- Department of Medical BiotechnologySchool of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
- ImmunologyAsthma & Allergy Research InstituteTehran University of Medical SciencesTehranIran
| | - Shadi Damough
- Department of Medical BiotechnologyBiotechnology Research CenterPasteur InstituteTehranIran
| | - Sara Mohseni
- Non‐metallic Materials Research GroupNiroo Research InstituteTehranIran
| | | | - Vahid Khakyzadeh
- Department of ChemistryK. N. Toosi University of TechnologyTehranIran
| | - Hamid Madanchi
- School of MedicineSemnan University of Medical SciencesSemnanIran
- Drug Design and Bioinformatics UnitDepartment of Medical BiotechnologyBiotechnology Research CenterPasteur Institute of IranTehranIran
| | - Gholam Ali Kardar
- Department of Medical BiotechnologySchool of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
- ImmunologyAsthma & Allergy Research InstituteTehran University of Medical SciencesTehranIran
| | - Payam Zarrintaj
- School of Chemical EngineeringOklahoma State UniversityStillwaterOklahomaUSA
| | - Mohammad Reza Saeb
- Department of Polymer TechnologyFaculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
227
|
Huang Z, Fu Z, Wang J. Review on Drug Regulatory Science Promoting COVID-19 Vaccine Development in China. ENGINEERING (BEIJING, CHINA) 2022; 10:127-132. [PMID: 35096437 PMCID: PMC8779850 DOI: 10.1016/j.eng.2022.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/12/2021] [Accepted: 01/06/2022] [Indexed: 05/27/2023]
Abstract
Regulatory science is a discipline that uses comprehensive methods of natural science, social science, and humanities to provide support for administrative decision-making through the development of new tools, standards, and approaches to assess the safety, efficacy, quality, and performance of regulated products. During the pandemics induced by infectious diseases, such as H1N1 flu, severe acute respiratory syndrome (SARS), and Middle East respiratory syndrome (MERS), regulatory science strongly supported the development of drugs and vaccines to respond to the viruses. In particular, with the support of research on drug regulatory science, vaccines have played a major role in the prevention and control of coronavirus disease 2019 (COVID-19). This review summarizes the overall state of the vaccine industry, research and development (R&D) of COVID-19 vaccines in China, and the general state of regulatory science and supervision for vaccines in China. Further, this review highlights how regulatory science has promoted the R&D of Chinese COVID-19 vaccines, with analyses from the aspects of national-level planning, relevant laws and regulations, technical guidelines, quality control platforms, and post-marketing supervision. Ultimately, this review provides a reference for the formulation of a vaccine development strategy in response to the current pandemic and the field of vaccine development in the post-pandemic era, as well as guidance on how to better respond to emerging and recurring infectious diseases that may occur in the future.
Collapse
Affiliation(s)
- Zhiming Huang
- National Medical Products Administration, Beijing 100053, China
| | - Zhihao Fu
- National Institutes for Food and Drug Control, Beijing 102629, China
| | - Junzhi Wang
- National Institutes for Food and Drug Control, Beijing 102629, China
| |
Collapse
|
228
|
Teo A, Chua CLL, Chan LLY. Airway models in a pandemic: Suitability of models in modeling SARS-CoV-2. PLoS Pathog 2022; 18:e1010432. [PMID: 35349597 PMCID: PMC8963546 DOI: 10.1371/journal.ppat.1010432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Louisa L. Y. Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
229
|
Martins M, Boggiatto PM, Buckley A, Cassmann ED, Falkenberg S, Caserta LC, Fernandes MHV, Kanipe C, Lager K, Palmer MV, Diel DG. From Deer-to-Deer: SARS-CoV-2 is efficiently transmitted and presents broad tissue tropism and replication sites in white-tailed deer. PLoS Pathog 2022; 18:e1010197. [PMID: 35312736 PMCID: PMC8970504 DOI: 10.1371/journal.ppat.1010197] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/31/2022] [Accepted: 03/10/2022] [Indexed: 01/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) in humans, has a broad host range, and is able to infect domestic and wild animal species. Notably, white-tailed deer (WTD, Odocoileus virginianus), the most widely distributed cervid species in the Americas, were shown to be highly susceptible to SARS-CoV-2 in challenge studies and reported natural infection/exposure rates approaching 30-40% in free-ranging WTD in the U.S. Thus, understanding the infection and transmission dynamics of SARS-CoV-2 in WTD is critical to prevent future zoonotic transmission to humans, at the human-WTD interface during hunting or venison farming, and for implementation of effective disease control measures. Here, we demonstrated that following intranasal inoculation with SARS-CoV-2 B.1 lineage, WTD fawns (~8-month-old) shed infectious virus up to day 5 post-inoculation (pi), with high viral loads shed in nasal and oral secretions. This resulted in efficient deer-to-deer transmission on day 3 pi. Consistent a with lack of infectious SARS-CoV-2 shedding after day 5 pi, no transmission was observed to contact animals added on days 6 and 9 pi. We have also investigated the tropism and sites of SARS-CoV-2 replication in adult WTD (3-4 years of age). Infectious virus was detected up to day 6 pi in nasal secretions, and from various respiratory-, lymphoid-, and central nervous system tissues, indicating broad tissue tropism and multiple sites of virus replication. The study provides important insights on the infection and transmission dynamics of SARS-CoV-2 in WTD, a wild animal species that is highly susceptible to infection and with the potential to become a reservoir for the virus in the field.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Paola M. Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Alexandra Buckley
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Eric D. Cassmann
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Shollie Falkenberg
- Ruminant Disease and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Leonardo C. Caserta
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Maureen H. V. Fernandes
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Carly Kanipe
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Kelly Lager
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Mitchell V. Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, United States of America
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
230
|
Forero‐Peña DA, Hernandez MM, Mozo Herrera IP, Collado Espinal IB, Páez Paz J, Ferro C, Flora‐Noda DM, Maricuto AL, Velásquez VL, Camejo‐Avila NA, Sordillo EM, Delgado‐Noguera LA, Perez‐Garcia LA, Morantes Rodríguez CG, Landaeta ME, Paniz‐Mondolfi AE. Remitting neuropsychiatric symptoms in COVID-19 patients: Viral cause or drug effect? J Med Virol 2022; 94:1154-1161. [PMID: 34755347 PMCID: PMC8661670 DOI: 10.1002/jmv.27443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023]
Abstract
Numerous reports of neuropsychiatric symptoms highlighted the pathologic potential of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its relationship the onset and/or exacerbation of mental disease. However, coronavirus disease 2019 (COVID-19) treatments, themselves, must be considered as potential catalysts for new-onset neuropsychiatric symptoms in COVID-19 patients. To date, immediate and long-term neuropsychiatric complications following SARS-CoV-2 infection are currently unknown. Here we report on five patients with SARS-CoV-2 infection with possible associated neuropsychiatric involvement, following them clinically until resolution of their symptoms. We will also discuss the contributory roles of chloroquine and dexamethasone in these neuropsychiatric presentations.
Collapse
Affiliation(s)
- David A. Forero‐Peña
- Department of Infectious DiseasesUniversity Hospital of CaracasCaracasVenezuela
- Department of Infectious DiseasesBiomedical Research and Therapeutic Vaccines InstituteCiudadBolivarVenezuela
| | - Matthew M. Hernandez
- Department of Pathology, Molecular and Cell‐based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | | | - Joselyn Páez Paz
- Psychiatry DepartmentUniversity Military Hospital ″Dr. Carlos Arvelo ″CaracasVenezuela
| | - Carlos Ferro
- Psychiatry DepartmentUniversity Hospital of CaracasCaracasVenezuela
| | - David M. Flora‐Noda
- Department of Infectious DiseasesUniversity Hospital of CaracasCaracasVenezuela
| | - Andrea L. Maricuto
- Department of Infectious DiseasesUniversity Hospital of CaracasCaracasVenezuela
| | - Viledy L. Velásquez
- Department of Infectious DiseasesUniversity Hospital of CaracasCaracasVenezuela
| | - Natasha A. Camejo‐Avila
- Department of Infectious DiseasesBiomedical Research and Therapeutic Vaccines InstituteCiudadBolivarVenezuela
| | - Emilia M. Sordillo
- Department of Pathology, Molecular and Cell‐based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Lourdes A. Delgado‐Noguera
- Instituto de Investigaciones Biomédicas IDB/Emerging Pathogens Network‐Incubadora Venezolana de la CienciaCabudareVenezuela
| | - Luis A. Perez‐Garcia
- Instituto de Investigaciones Biomédicas IDB/Emerging Pathogens Network‐Incubadora Venezolana de la CienciaCabudareVenezuela
| | | | | | - Alberto E. Paniz‐Mondolfi
- Department of Pathology, Molecular and Cell‐based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
231
|
Upadhya S, Rehman J, Malik AB, Chen S. Mechanisms of Lung Injury Induced by SARS-CoV-2 Infection. Physiology (Bethesda) 2022; 37:88-100. [PMID: 34698589 PMCID: PMC8873036 DOI: 10.1152/physiol.00033.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
The lung is the major target organ of SARS-CoV-2 infection, which causes COVID-19. Here, we outline the multistep mechanisms of lung epithelial and endothelial injury induced by SARS-CoV-2: direct viral infection, chemokine/cytokine-mediated damage, and immune cell-mediated lung injury. Finally, we discuss the recent progress in terms of antiviral therapeutics as well as the development of anti-inflammatory or immunomodulatory therapeutic approaches. This review also provides a systematic overview of the models for studying SARS-CoV-2 infection and discusses how an understanding of mechanisms of lung injury will help identify potential targets for future drug development to mitigate lung injury.
Collapse
Affiliation(s)
- Samsara Upadhya
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois
| | - Asrar B Malik
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, New York
| |
Collapse
|
232
|
Halfmann PJ, Iida S, Iwatsuki-Horimoto K, Maemura T, Kiso M, Scheaffer SM, Darling TL, Joshi A, Loeber S, Singh G, Foster SL, Ying B, Case JB, Chong Z, Whitener B, Moliva J, Floyd K, Ujie M, Nakajima N, Ito M, Wright R, Uraki R, Warang P, Gagne M, Li R, Sakai-Tagawa Y, Liu Y, Larson D, Osorio JE, Hernandez-Ortiz JP, Henry AR, Ciuoderis K, Florek KR, Patel M, Odle A, Wong LYR, Bateman AC, Wang Z, Edara VV, Chong Z, Franks J, Jeevan T, Fabrizio T, DeBeauchamp J, Kercher L, Seiler P, Gonzalez-Reiche AS, Sordillo EM, Chang LA, van Bakel H, Simon V, Douek DC, Sullivan NJ, Thackray LB, Ueki H, Yamayoshi S, Imai M, Perlman S, Webby RJ, Seder RA, Suthar MS, García-Sastre A, Schotsaert M, Suzuki T, Boon ACM, Diamond MS, Kawaoka Y. SARS-CoV-2 Omicron virus causes attenuated disease in mice and hamsters. Nature 2022; 603:687-692. [PMID: 35062015 PMCID: PMC8942849 DOI: 10.1038/s41586-022-04441-6] [Citation(s) in RCA: 454] [Impact Index Per Article: 151.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/19/2022] [Indexed: 11/09/2022]
Abstract
The recent emergence of B.1.1.529, the Omicron variant1,2, has raised concerns of escape from protection by vaccines and therapeutic antibodies. A key test for potential countermeasures against B.1.1.529 is their activity in preclinical rodent models of respiratory tract disease. Here, using the collaborative network of the SARS-CoV-2 Assessment of Viral Evolution (SAVE) programme of the National Institute of Allergy and Infectious Diseases (NIAID), we evaluated the ability of several B.1.1.529 isolates to cause infection and disease in immunocompetent and human ACE2 (hACE2)-expressing mice and hamsters. Despite modelling data indicating that B.1.1.529 spike can bind more avidly to mouse ACE2 (refs. 3,4), we observed less infection by B.1.1.529 in 129, C57BL/6, BALB/c and K18-hACE2 transgenic mice than by previous SARS-CoV-2 variants, with limited weight loss and lower viral burden in the upper and lower respiratory tracts. In wild-type and hACE2 transgenic hamsters, lung infection, clinical disease and pathology with B.1.1.529 were also milder than with historical isolates or other SARS-CoV-2 variants of concern. Overall, experiments from the SAVE/NIAID network with several B.1.1.529 isolates demonstrate attenuated lung disease in rodents, which parallels preliminary human clinical data.
Collapse
Affiliation(s)
- Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Tadashi Maemura
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Maki Kiso
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Astha Joshi
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Samantha Loeber
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephanie L Foster
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Baoling Ying
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Bradley Whitener
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Juan Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katharine Floyd
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Michiko Ujie
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Noriko Nakajima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mutsumi Ito
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ryan Wright
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rong Li
- Department of Animal Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, USA
| | - Yuko Sakai-Tagawa
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yanan Liu
- Department of Animal Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, USA
| | - Deanna Larson
- Department of Animal Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, USA
| | - Jorge E Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
- Colombia/Wisconsin One-Health Consortium and One-Health Genomic Laboratory, Universidad Nacional de Colombia, Medellín, Colombia
| | - Juan P Hernandez-Ortiz
- Colombia/Wisconsin One-Health Consortium and One-Health Genomic Laboratory, Universidad Nacional de Colombia, Medellín, Colombia
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Karl Ciuoderis
- Colombia/Wisconsin One-Health Consortium and One-Health Genomic Laboratory, Universidad Nacional de Colombia, Medellín, Colombia
| | | | - Mit Patel
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Abby Odle
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lok-Yin Roy Wong
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | | | - Zhongde Wang
- Department of Animal Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, USA
| | - Venkata-Viswanadh Edara
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - John Franks
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Trushar Jeevan
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Thomas Fabrizio
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jennifer DeBeauchamp
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lisa Kercher
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Patrick Seiler
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Emilia Mia Sordillo
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Hiroshi Ueki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mehul S Suthar
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
| |
Collapse
|
233
|
Pérez P, Lázaro-Frías A, Zamora C, Sánchez-Cordón PJ, Astorgano D, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. A Single Dose of an MVA Vaccine Expressing a Prefusion-Stabilized SARS-CoV-2 Spike Protein Neutralizes Variants of Concern and Protects Mice From a Lethal SARS-CoV-2 Infection. Front Immunol 2022; 12:824728. [PMID: 35154086 PMCID: PMC8829548 DOI: 10.3389/fimmu.2021.824728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 12/23/2022] Open
Abstract
We generated an optimized COVID-19 vaccine candidate based on the modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein, termed MVA-CoV2-S(3P). The S(3P) protein was expressed at higher levels (2-fold) than the non-stabilized S in cells infected with the corresponding recombinant MVA viruses. One single dose of MVA-CoV2-S(3P) induced higher IgG and neutralizing antibody titers against parental SARS-CoV-2 and variants of concern than MVA-CoV2-S in wild-type C57BL/6 and in transgenic K18-hACE2 mice. In immunized C57BL/6 mice, two doses of MVA-CoV2-S or MVA-CoV2-S(3P) induced similar levels of SARS-CoV-2-specific B- and T-cell immune responses. Remarkably, a single administration of MVA-CoV2-S(3P) protected all K18-hACE2 mice from morbidity and mortality caused by SARS-CoV-2 infection, reducing SARS-CoV-2 viral loads, histopathological lesions, and levels of pro-inflammatory cytokines in the lungs. These results demonstrated that expression of a novel full-length prefusion-stabilized SARS-CoV-2 S protein by the MVA poxvirus vector enhanced immunogenicity and efficacy against SARS-CoV-2 in animal models, further supporting MVA-CoV2-S(3P) as an optimized vaccine candidate for clinical trials.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro J Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - José M Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
234
|
Microglia Do Not Restrict SARS-CoV-2 Replication following Infection of the Central Nervous System of K18-Human ACE2 Transgenic Mice. J Virol 2022; 96:e0196921. [PMID: 34935438 PMCID: PMC8865461 DOI: 10.1128/jvi.01969-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Unlike SARS-CoV-1 and MERS-CoV, infection with SARS-CoV-2, the viral pathogen responsible for COVID-19, is often associated with neurologic symptoms that range from mild to severe, yet increasing evidence argues the virus does not exhibit extensive neuroinvasive properties. We demonstrate SARS-CoV-2 can infect and replicate in human iPSC-derived neurons and that infection shows limited antiviral and inflammatory responses but increased activation of EIF2 signaling following infection as determined by RNA sequencing. Intranasal infection of K18 human ACE2 transgenic mice (K18-hACE2) with SARS-CoV-2 resulted in lung pathology associated with viral replication and immune cell infiltration. In addition, ∼50% of infected mice exhibited CNS infection characterized by wide-spread viral replication in neurons accompanied by increased expression of chemokine (Cxcl9, Cxcl10, Ccl2, Ccl5 and Ccl19) and cytokine (Ifn-λ and Tnf-α) transcripts associated with microgliosis and a neuroinflammatory response consisting primarily of monocytes/macrophages. Microglia depletion via administration of colony-stimulating factor 1 receptor inhibitor, PLX5622, in SARS-CoV-2 infected mice did not affect survival or viral replication but did result in dampened expression of proinflammatory cytokine/chemokine transcripts and a reduction in monocyte/macrophage infiltration. These results argue that microglia are dispensable in terms of controlling SARS-CoV-2 replication in in the K18-hACE2 model but do contribute to an inflammatory response through expression of pro-inflammatory genes. Collectively, these findings contribute to previous work demonstrating the ability of SARS-CoV-2 to infect neurons as well as emphasizing the potential use of the K18-hACE2 model to study immunological and neuropathological aspects related to SARS-CoV-2-induced neurologic disease. IMPORTANCE Understanding the immunological mechanisms contributing to both host defense and disease following viral infection of the CNS is of critical importance given the increasing number of viruses that are capable of infecting and replicating within the nervous system. With this in mind, the present study was undertaken to evaluate the role of microglia in aiding in host defense following experimental infection of the central nervous system (CNS) of K18-hACE2 with SARS-CoV-2, the causative agent of COVID-19. Neurologic symptoms that range in severity are common in COVID-19 patients and understanding immune responses that contribute to restricting neurologic disease can provide important insight into better understanding consequences associated with SARS-CoV-2 infection of the CNS.
Collapse
|
235
|
Jawalagatti V, Kirthika P, Hewawaduge C, Park JY, Yang MS, Oh B, So MY, Kim B, Lee JH. A Simplified SARS-CoV-2 Mouse Model Demonstrates Protection by an Oral Replicon-Based mRNA Vaccine. Front Immunol 2022; 13:811802. [PMID: 35250985 PMCID: PMC8888445 DOI: 10.3389/fimmu.2022.811802] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/25/2022] [Indexed: 12/24/2022] Open
Abstract
A mouse model of SARS-CoV-2 that can be developed in any molecular biology lab with standard facilities will be valuable in evaluating drugs and vaccines. Here we present a simplified SARS-CoV-2 mouse model exploiting the rapid adenoviral purification method. Mice that are sensitive to SARS-CoV-2 infection were generated by transducing human angiotensin-converting enzyme 2 (hACE2) by an adenovirus. The expression kinetics of the hACE2 in transduced mice were assessed by immunohistochemistry, RT-PCR, and qPCR. Further, the ability of the hACE2 to support viral replication was determined in vitro and in vivo. The hACE2 expression in the lungs of mice was observed for at least nine days after transduction. The murine macrophages expressing hACE2 supported viral replication with detection of high viral titers. Next, in vivo studies were carried out to determine viral replication and lung disease following SARS-CoV-2 challenge. The model supported viral replication, and the challenged mouse developed lung disease characteristic of moderate interstitial pneumonia. Further, we illustrated the utility of the system by demonstrating protection using an oral mRNA vaccine. The multicistronic vaccine design enabled by the viral self-cleaving peptides targets receptor binding domain (RBD), heptad repeat domain (HR), membrane glycoprotein (M) and epitopes of nsp13 of parental SARS-CoV-2. Further, Salmonella and Semliki Forest virus replicon were exploited, respectively, for gene delivery and mRNA expression. We recorded potent cross-protective neutralizing antibodies in immunized mice against the SARS-CoV-2 delta variant. The vaccine protected the mice against viral replication and SARS-CoV-2-induced weight loss and lung pathology. The findings support the suitability of the model for preclinical evaluation of anti-SARS-CoV-2 therapies and vaccines. In addition, the findings provide novel insights into mRNA vaccine design against infectious diseases not limiting to SARS-CoV-2.
Collapse
Affiliation(s)
- Vijayakumar Jawalagatti
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Perumalraja Kirthika
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Ji-Young Park
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Myeon-Sik Yang
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Byungkwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Mi Young So
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Bumseok Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
236
|
Lázaro-Frías A, Pérez P, Zamora C, Sánchez-Cordón PJ, Guzmán M, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. Full efficacy and long-term immunogenicity induced by the SARS-CoV-2 vaccine candidate MVA-CoV2-S in mice. NPJ Vaccines 2022; 7:17. [PMID: 35140227 PMCID: PMC8828760 DOI: 10.1038/s41541-022-00440-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
Two doses of the MVA-CoV2-S vaccine candidate expressing the SARS-CoV-2 spike (S) protein protected K18-hACE2 transgenic mice from a lethal dose of SARS-CoV-2. This vaccination regimen prevented virus replication in the lungs, reduced lung pathology, and diminished levels of pro-inflammatory cytokines. High titers of IgG antibodies against S and receptor-binding domain (RBD) proteins and of neutralizing antibodies were induced against parental virus and variants of concern, markers that correlated with protection. Similar SARS-CoV-2-specific antibody responses were observed at prechallenge and postchallenge in the two-dose regimen, while the single-dose treatment does not avoid vaccine breakthrough infection. All vaccinated animals survived infection and were also protected to SARS-CoV-2 reinfection. Furthermore, two MVA-CoV2-S doses induced long-term memory S-specific humoral and cellular immune responses in C57BL/6 mice, 6 months after immunization. The efficacy and immunological benefits of the MVA-CoV2-S vaccine candidate against COVID-19 supports its consideration for human clinical trials.
Collapse
Affiliation(s)
- Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Pedro J Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), 28130, Valdeolmos, Madrid, Spain
| | - María Guzmán
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), 28041, Madrid, Spain.,Universidad Complutense School of Medicine, 28040, Madrid, Spain
| | - José M Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
237
|
Yan F, Li E, Wang T, Li Y, Liu J, Wang W, Qin T, Su R, Pei H, Wang S, Feng N, Zhao Y, Yang S, Xia X, Gao Y. Characterization of Two Heterogeneous Lethal Mouse-Adapted SARS-CoV-2 Variants Recapitulating Representative Aspects of Human COVID-19. Front Immunol 2022; 13:821664. [PMID: 35197985 PMCID: PMC8858946 DOI: 10.3389/fimmu.2022.821664] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/13/2022] [Indexed: 12/15/2022] Open
Abstract
New emerging severe acute respiratory syndrome 2 (SARS-CoV-2) has caused a worldwide pandemic. Several animal models of coronavirus disease 2019 (COVID-19) have been developed and applied to antiviral research. In this study, two lethal mouse-adapted SARS-CoV-2 variants (BMA8 and C57MA14) with different virulence were generated from different hosts, which are characterized by high viral replication titers in the upper and lower respiratory tract, pulmonary pathology, cytokine storm, cellular tropism, lymphopenia, and neutrophilia. Two variants exhibit host genetics-related and age-dependent morbidity and mortality in mice, exquisitely reflecting the clinical manifestation of asymptomatic, moderate, and severe COVID-19 patients. Notably, both variants equally weaken the neutralization capacity of the serum derived from COVID-19 convalescent, but the C57MA14 variant showed a much higher virulence than the BMA8 variant in vitro. Q489H substitution in the receptor-binding domain (RBD) of BMA8 and C57MA14 variants results in the receptors of SARS-CoV-2 switching from human angiotensin-converting enzyme 2 (hACE2) to murine angiotensin-converting enzyme 2 (mACE2). Additionally, A22D and A36V mutation in E protein were first reported in our study, which potentially contributed to the virulence difference between the two variants. Of note, the protective efficacy of the novel bacterium-like particle (BLP) vaccine candidate was validated using the BMA8- or C57MA14-infected aged mouse model. The BMA8 variant- and C57MA14 variant-infected models provide a relatively inexpensive and accessible evaluation platform for assessing the efficacy of vaccines and novel therapeutic approaches. This will promote further research in the transmissibility and pathogenicity mechanisms of SARS-CoV-2.
Collapse
Affiliation(s)
- Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Entao Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuanguo Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jun Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tian Qin
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- School of Life Sciences, Northeast Normal University, Changchun, China
| | - Rina Su
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Hongyan Pei
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- *Correspondence: Yuwei Gao,
| |
Collapse
|
238
|
Barroso R, Vieira-Pires A, Antunes A, Fidalgo-Carvalho I. Susceptibility of Pets to SARS-CoV-2 Infection: Lessons from a Seroepidemiologic Survey of Cats and Dogs in Portugal. Microorganisms 2022; 10:345. [PMID: 35208799 PMCID: PMC8879010 DOI: 10.3390/microorganisms10020345] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Betacoronavirus (β-CoV) are positive single-stranded RNA viruses known to infect mammals. In 2019, a novel zoonotic β-CoV emerged, the severe acute respiratory syndrome (SARS)-CoV-2. Although the most frequent SARS-CoV-2 transmission route is within humans, spillover from humans to domestic and wild animals has been reported, including cats (Felis catus), dogs (Canis lupus familiaris), and minks (Neovision vision). In order to understand the potential role of domestic animals in SARS-CoV-2 global transmission, as well their susceptibility to infection, a seroepidemiologic survey of cats and dogs in Portugal was conducted. Antibodies against SARS-CoV-2 were detected in 15/69 (21.74%) cats and 7/148 (4.73%) dogs. Of the SARS-CoV-2 seropositive animals, 11/22 (50.00%) were possibly infected by human-to-animal transmission, and 5/15 (33.33%) cats were probably infected by cat-to-cat transmission. Moreover, one dog tested positive for SARS-CoV-2 RNA. Data suggest that cats and dogs are susceptible to SARS-CoV-2 infection in natural conditions. Hence, a one-health approach is crucial in the SARS-CoV-2 pandemic to understand the risk factors beyond infection in a human-animal environment interface.
Collapse
Affiliation(s)
- Ricardo Barroso
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal;
| | - Alexandre Vieira-Pires
- Equigerminal, S.A., Rua Eduardo Correia, n°13 lote 20.12, 3030-507 Coimbra, Portugal; (A.V.-P.); (I.F.-C.)
- CNC-Center for Neuroscience and Cell Biology, CIBB, PhD Programme in Experimental Biology and Biomedicine (PDBEB) and Institute for Interdisciplinary Research, University of Coimbra (III UC), Rua Larga, 3004-504 Coimbra, Portugal
| | - Agostinho Antunes
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal;
- CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450-208 Porto, Portugal
| | - Isabel Fidalgo-Carvalho
- Equigerminal, S.A., Rua Eduardo Correia, n°13 lote 20.12, 3030-507 Coimbra, Portugal; (A.V.-P.); (I.F.-C.)
- CIVG - Vasco da Gama Research Center, Escola Universitária Vasco da Gama (EUVG), Campus Universitário, Av. José R. Sousa Fernandes, 3020-210 Coimbra, Portugal
| |
Collapse
|
239
|
Da Costa CBP, Cruz ACDM, Penha JCQ, Castro HC, Da Cunha LER, Ratcliffe NA, Cisne R, Martins FJ. Using in vivo animal models for studying SARS-CoV-2. Expert Opin Drug Discov 2022; 17:121-137. [PMID: 34727803 PMCID: PMC8567288 DOI: 10.1080/17460441.2022.1995352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The search for an animal model capable of reproducing the physiopathology of the COVID-19, and also suitable for evaluating the efficacy and safety of new drugs has become a challenge for many researchers. AREAS COVERED This work reviews the current animal models for in vivo tests with SARS-CoV-2 as well as the challenges involved in the safety and efficacy trials. EXPERT OPINION Studies have reported the use of nonhuman primates, ferrets, mice, Syrian hamsters, lagomorphs, mink, and zebrafish in experiments that aimed to understand the course of COVID-19 or test vaccines and other drugs. In contrast, the assays with animal hyperimmune sera have only been used in in vitro assays. Finding an animal that faithfully reproduces all the characteristics of the disease in humans is difficult. Some models may be more complex to work with, such as monkeys, or require genetic manipulation so that they can express the human ACE2 receptor, as in the case of mice. Although some models are more promising, possibly the use of more than one animal model represents the best scenario. Therefore, further studies are needed to establish an ideal animal model to help in the development of other treatment strategies besides vaccines.
Collapse
Affiliation(s)
- Camila B. P. Da Costa
- Technological Development and Innovation Laboratory of the Industrial Board, Instituto Vital Brazil, Rio De Janeiro, Brazil
- Programa de Pós-graduação em Ciências e Biotecnologia, IB, UFF, Rio de Janeiro, Brazil
| | | | - Julio Cesar Q Penha
- Programa de Pós-graduação em Ciências e Biotecnologia, IB, UFF, Rio de Janeiro, Brazil
| | - Helena C Castro
- Programa de Pós-graduação em Ciências e Biotecnologia, IB, UFF, Rio de Janeiro, Brazil
| | - Luis E. R. Da Cunha
- Technological Development and Innovation Laboratory of the Industrial Board, Instituto Vital Brazil, Rio De Janeiro, Brazil
| | - Norman A Ratcliffe
- Programa de Pós-graduação em Ciências e Biotecnologia, IB, UFF, Rio de Janeiro, Brazil
- Department of Biociences, College of Science, Swansea University, Swansea, UK
| | - Rafael Cisne
- Programa de Pós-graduação em Ciências e Biotecnologia, IB, UFF, Rio de Janeiro, Brazil
| | | |
Collapse
|
240
|
Lu J, Yin Q, Pei R, Zhang Q, Qu Y, Pan Y, Sun L, Gao D, Liang C, Yang J, Wu W, Li J, Cui Z, Wang Z, Li X, Li D, Wang S, Duan K, Guan W, Liang M, Yang X. Nasal delivery of broadly neutralizing antibodies protects mice from lethal challenge with SARS-CoV-2 delta and omicron variants. Virol Sin 2022; 37:238-247. [PMID: 35527227 PMCID: PMC8855614 DOI: 10.1016/j.virs.2022.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
Multiple new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have constantly emerged, as the delta and omicron variants, which have developed resistance to currently gained neutralizing antibodies. This highlights a critical need to discover new therapeutic agents to overcome the variants mutations. Despite the availability of vaccines against coronavirus disease 2019 (COVID-19), the use of broadly neutralizing antibodies has been considered as an alternative way for the prevention or treatment of SARS-CoV-2 variants infection. Here, we show that the nasal delivery of two previously characterized broadly neutralizing antibodies (F61 and H121) protected K18-hACE2 mice against lethal challenge with SARS-CoV-2 variants. The broadly protective efficacy of the F61 or F61/F121 cocktail antibodies was evaluated by lethal challenge with the wild strain (WIV04) and multiple variants, including beta (B.1.351), delta (B.1.617.2), and omicron (B.1.1.529) at 200 or 1000 TCID50, and the minimum antibody administration doses (5–1.25 mg/kg body weight) were also evaluated with delta and omicron challenge. Fully prophylactic protections were found in all challenged groups with both F61 and F61/H121 combination at the administration dose of 20 mg/kg body weight, and corresponding mice lung viral RNA showed negative, with almost all alveolar septa and cavities remaining normal. Furthermore, low-dose antibody treatment induced significant prophylactic protection against lethal challenge with delta and omicron variants, whereas the F61/H121 combination showed excellent results against omicron infection. Our findings indicated the potential use of broadly neutralizing monoclonal antibodies as prophylactic and therapeutic agent for protection of current emerged SARS-CoV-2 variants infection. The mAbs could be detected in lungs shortly after nasal spray and kept in the lungs for a long time. High dose mAbs nasal delivery could fully prophylactic protection mice from omicron lethal challenged. Significant enhancement of broadly neutralizing activity against variants were confirmed in F61 and H121 combination use.
Collapse
|
241
|
Kulkarni PG, Sakharkar A, Banerjee T. Understanding the role of nACE2 in neurogenic hypertension among COVID-19 patients. Hypertens Res 2022; 45:254-269. [PMID: 34848886 PMCID: PMC8630198 DOI: 10.1038/s41440-021-00800-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/20/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
Currently, the third and fourth waves of the coronavirus disease -19 (COVID-19) pandemic are creating havoc in many parts of the world. Although vaccination programs have been launched in most countries, emerging new strains of the virus along with geographical variations are leading to varying success rates of the available vaccines. The presence of comorbidities such as diabetes, cardiovascular diseases and hypertension is responsible for increasing the severity of COVID-19 and, thus, the COVID-19 mortality rate. Angiotensin-converting enzyme 2 (ACE2), which is utilized by SARS-CoV-2 for entry into host cells, is widely expressed in the lungs, kidneys, testes, gut, adipose tissue, and brain. Infection within host cells mediates RAS overactivation, which leads to a decrease in the ACE2/ACE ratio, AT2R/AT1R ratio, and MasR/AT1R ratio. Such imbalances lead to the development of heightened inflammatory responses, such as cytokine storms, leading to post-COVID-19 complications and mortality. As the association of SARS-CoV-2 infection and hypertension remains unclear, this report provides an overview of the effects of SARS-CoV-2 infection on patients with hypertension. We discuss here the interaction of ACE2 with SARS-CoV-2, focusing on neuronal ACE2 (nACE2), and further shed light on the possible involvement of nACE2 in hypertension. SARS-CoV-2 enters the brain through neuronal ACE2 and spreads in various regions of the brain. The effect of viral binding to neuronal ACE2 in areas of the brain that regulate salt/water balance and blood pressure is also discussed in light of the neural regulation of hypertension in COVID-19.
Collapse
Affiliation(s)
- Prakash G Kulkarni
- Department of Biotechnology, Savitribai Phule Pune University Ganeshkhind Road, Pune, 411007, India
| | - Amul Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University Ganeshkhind Road, Pune, 411007, India.
| | - Tanushree Banerjee
- Molecular Neuroscience Research Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth Survey No 87/88, Mumbai Bangalore Express Highway, Tathawade, Pune, 411 033, India.
| |
Collapse
|
242
|
The impact of COVID-19 on the comorbidities: A review of recent updates for combating it. Saudi J Biol Sci 2022; 29:3586-3599. [PMID: 35165505 PMCID: PMC8828435 DOI: 10.1016/j.sjbs.2022.02.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 can also affect organs other than the lungs, including the brain, heart, and gastrointestinal system. Patients with Cancer, HIV, COPD, neurological, and CVDs are more prone to the COVID-19 associated complications, leading to a drastic rise in morbidity and mortality. Elderly and pre-existing polypharmacy patients have worsened COVID-19 associated complications. When a person with comorbidity is infected with SARS-CoV-2, it becomes more dangerous, and managing these patients with adequate medical care is critical to their survival. A co-morbid person should adhere to preventive measures to reduce mortality, including regular handwashing with soap or using an alcohol-based hand sanitizer, minimizing in person contact and practicing social distance, wearing a face mask in public places, and avoiding going to public places unless essential are among the precautional measures.
Coronavirus disease is caused by the SARS-CoV-2 virus. The virus first appeared in Wuhan (China) in December 2019 and has spread globally. Till now, it affected 269 million people with 5.3 million deaths in 224 countries and territories. With the emergence of variants like Omicron, the COVID-19 cases grew exponentially, with thousands of deaths. The general symptoms of COVID-19 include fever, sore throat, cough, lung infections, and, in severe cases, acute respiratory distress syndrome, sepsis, and death. SARS-CoV-2 predominantly affects the lung, but it can also affect other organs such as the brain, heart, and gastrointestinal system. It is observed that 75 % of hospitalized COVID-19 patients have at least one COVID-19 associated comorbidity. The most common reported comorbidities are hypertension, NDs, diabetes, cancer, endothelial dysfunction, and CVDs. Moreover, older and pre-existing polypharmacy patients have worsened COVID-19 associated complications. SARS-CoV-2 also results in the hypercoagulability issues like gangrene, stroke, pulmonary embolism, and other associated complications. This review aims to provide the latest information on the impact of the COVID-19 on pre-existing comorbidities such as CVDs, NDs, COPD, and other complications. This review will help us to understand the current scenario of COVID-19 and comorbidities; thus, it will play an important role in the management and decision-making efforts to tackle such complications.
Collapse
|
243
|
Fumagalli V, Ravà M, Marotta D, Di Lucia P, Laura C, Sala E, Grillo M, Bono E, Giustini L, Perucchini C, Mainetti M, Sessa A, Garcia-Manteiga JM, Donnici L, Manganaro L, Delbue S, Broccoli V, De Francesco R, D’Adamo P, Kuka M, Guidotti LG, Iannacone M. Administration of aerosolized SARS-CoV-2 to K18-hACE2 mice uncouples respiratory infection from fatal neuroinvasion. Sci Immunol 2022; 7:eabl9929. [PMID: 34812647 PMCID: PMC9835999 DOI: 10.1126/sciimmunol.abl9929] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The development of a tractable small animal model faithfully reproducing human coronavirus disease 2019 pathogenesis would arguably meet a pressing need in biomedical research. Thus far, most investigators have used transgenic mice expressing the human ACE2 in epithelial cells (K18-hACE2 transgenic mice) that are intranasally instilled with a liquid severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) suspension under deep anesthesia. Unfortunately, this experimental approach results in disproportionate high central nervous system infection leading to fatal encephalitis, which is rarely observed in humans and severely limits this model’s usefulness. Here, we describe the use of an inhalation tower system that allows exposure of unanesthetized mice to aerosolized virus under controlled conditions. Aerosol exposure of K18-hACE2 transgenic mice to SARS-CoV-2 resulted in robust viral replication in the respiratory tract, anosmia, and airway obstruction but did not lead to fatal viral neuroinvasion. When compared with intranasal inoculation, aerosol infection resulted in a more pronounced lung pathology including increased immune infiltration, fibrin deposition, and a transcriptional signature comparable to that observed in SARS-CoV-2–infected patients. This model may prove useful for studies of viral transmission, disease pathogenesis (including long-term consequences of SARS-CoV-2 infection), and therapeutic interventions.
Collapse
Affiliation(s)
- Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Micol Ravà
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Davide Marotta
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Laura
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy.,Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Eleonora Sala
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Marta Grillo
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elisa Bono
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Perucchini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marta Mainetti
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandro Sessa
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Lorena Donnici
- INGM - Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, Milan, Italy
| | - Lara Manganaro
- INGM - Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, Milan, Italy
| | - Serena Delbue
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Vania Broccoli
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,National Research Council of Italy, Institute of Neuroscience
| | - Raffaele De Francesco
- INGM - Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, Milan, Italy,Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Italy
| | - Patrizia D’Adamo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Center of Advanced Services for in-vivo testing – Animal behavior Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mirela Kuka
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Luca G. Guidotti
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy.,Correspondence to: or
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy.,Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Correspondence to: or
| |
Collapse
|
244
|
Bai Y, Wang Q, Liu M, Bian L, Liu J, Gao F, Mao Q, Wang Z, Wu X, Xu M, Liang Z. The next major emergent infectious disease: reflections on vaccine emergency development strategies. Expert Rev Vaccines 2022; 21:471-481. [PMID: 35080441 DOI: 10.1080/14760584.2022.2027240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Major emergent infectious diseases (MEID) pose the most serious threat to human health. The research proposes targeted response strategies for the prevention and control of potential MEID. AREAS COVERED Based on the analysis of infectious diseases, this research analyzes pandemics that have a high probability of occurrence and aims to synthesize the past experience and lessons learned of controlling infectious diseases such as coronavirus, influenza, Ebola, etc. In addition, by integrating major infectious disease response guidelines developed by WHO, the European Union, the United States, and the United Kingdom, we intend to bring forward national vaccine R&D development strategies for emergency use. EXPERT OPINION We advise to establish and improve existing laws, regulations, and also prevention and control systems for the emergent R&D and application of vaccines in response to potential infectious diseases. The strategies would not only help increase the various abilities in response to the research, development, evaluation, production, and supervision of emergency vaccines, but also establish surrogate endpoint of immunogenicity protection in early clinical studies to enable a rapid evaluation of the efficacy of emergency vaccines.
Collapse
Affiliation(s)
- Yu Bai
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Qian Wang
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Mingchen Liu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Lianlian Bian
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Jianyang Liu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Fan Gao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Qunying Mao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Zhongfang Wang
- Guangzhou Laboratory. No. 9 XingDaoHuanBei Road, Guangzhou, China
| | - Xing Wu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Miao Xu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Zhenglun Liang
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| |
Collapse
|
245
|
Saravanan UB, Namachivayam M, Jeewon R, Huang JD, Durairajan SSK. Animal models for SARS-CoV-2 and SARS-CoV-1 pathogenesis, transmission and therapeutic evaluation. World J Virol 2022; 11:40-56. [PMID: 35117970 PMCID: PMC8788210 DOI: 10.5501/wjv.v11.i1.40] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/22/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023] Open
Abstract
There is a critical need to develop animal models to alleviate vaccine and drug development difficulties against zoonotic viral infections. The coronavirus family, which includes severe acute respiratory syndrome coronavirus 1 and severe acute respiratory syndrome coronavirus 2, crossed the species barrier and infected humans, causing a global outbreak in the 21st century. Because humans do not have pre-existing immunity against these viral infections and with ethics governing clinical trials, animal models are therefore being used in clinical studies to facilitate drug discovery and testing efficacy of vaccines. The ideal animal models should reflect the viral replication, clinical signs, and pathological responses observed in humans. Different animal species should be tested to establish an appropriate animal model to study the disease pathology, transmission and evaluation of novel vaccine and drug candidates to treat coronavirus disease 2019. In this context, the present review summarizes the recent progress in developing animal models for these two pathogenic viruses and highlights the utility of these models in studying SARS-associated coronavirus diseases.
Collapse
Affiliation(s)
- Udhaya Bharathy Saravanan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Mayurikaa Namachivayam
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Rajesh Jeewon
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Reduit 80837, Mauritius
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong Province, China
| | | |
Collapse
|
246
|
Zhu G, Du S, Wang Y, Huang X, Hu G, Lu X, Li D, Zhu Y, Qu D, Cai Q, Liu L, Du M. Delayed Antiviral Immune Responses in Severe Acute Respiratory Syndrome Coronavirus Infected Pregnant Mice. Front Microbiol 2022; 12:806902. [PMID: 35126335 PMCID: PMC8814454 DOI: 10.3389/fmicb.2021.806902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Sex differences in immune responses had been reported to correlate with different symptoms and mortality in the disease course of coronavirus disease 2019 (COVID-19). However, whether severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection interferes with females’ fertility and causes different symptoms among pregnant and non-pregnant females remains unknown. Here, we examined the differences in viral loads, SARS-CoV-2-specific antibody titers, proinflammatory cytokines, and levels of T cell activation after SARS-CoV-2 sub-lethal infection between pregnant and non-pregnant human Angiotensin-Converting Enzyme II (ACE2) transgenic mouse models. Both mice showed elevated levels of viral loads in the lung at 4 days post-infection (dpi). However, viral loads in the pregnant group remained elevated at 7 dpi while decreased in the non-pregnant group. Consistent with viral loads, increased production of proinflammatory cytokines was detected from the pregnant group, and the IgM or SARS-CoV-2-specific IgG antibody in serum of pregnant mice featured delayed elevation compared with non-pregnant mice. Moreover, by accessing kinetics of activation marker expression of peripheral T cells after infection, a lower level of CD8+ T cell activation was observed in pregnant mice, further demonstrating the difference of immune-response between pregnant and non-pregnant mice. Although vertical transmission did not occur as SARS-CoV-2 RNA was absent in the uterus and fetus from the infected pregnant mice, a lower pregnancy rate was observed when the mice were infected before embryo implantation after mating, indicating that SARS-CoV-2 infection may interfere with mice’s fertility at a specific time window. In summary, pregnant mice bear a weaker ability to eliminate the SARS-CoV-2 virus than non-pregnant mice, which was correlated with lower levels of antibody production and T cell activation.
Collapse
Affiliation(s)
- Guohua Zhu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shujuan Du
- MOE & NHC & CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuyan Wang
- MOE & NHC & CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xixi Huang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Gaowei Hu
- MOE & NHC & CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Lu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yizhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Di Qu
- MOE & NHC & CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiliang Cai
- MOE & NHC & CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- Qiliang Cai,
| | - Lu Liu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- Lu Liu,
| | - Meirong Du
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Meirong Du,
| |
Collapse
|
247
|
Dolskiy AA, Gudymo AS, Taranov OS, Grishchenko IV, Shitik EM, Prokopov DY, Soldatov VO, Sobolevskaya EV, Bodnev SA, Danilchenko NV, Moiseeva AA, Torzhkova PY, Bulanovich YA, Onhonova GS, Ivleva EK, Kubekina MV, Belykh AE, Tregubchak TV, Ryzhikov AB, Gavrilova EV, Maksyutov RA, Deykin AV, Yudkin DV. The Tissue Distribution of SARS-CoV-2 in Transgenic Mice With Inducible Ubiquitous Expression of hACE2. Front Mol Biosci 2022; 8:821506. [PMID: 35118120 PMCID: PMC8804232 DOI: 10.3389/fmolb.2021.821506] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
The novel coronavirus disease COVID-19 has become one of the most socially significant infections. One of the main models for COVID-19 pathogenesis study and anti-COVID-19 drug development is laboratory animals sensitive to the virus. Herein, we report SARS-CoV-2 infection in novel transgenic mice conditionally expressing human ACE2 (hACE2), with a focus on viral distribution after intranasal inoculation. Transgenic mice carrying hACE2 under the floxed STOP cassette [(hACE2-LoxP(STOP)] were mated with two types of Cre-ERT2 strains (UBC-Cre and Rosa-Cre). The resulting offspring with temporal control of transgene expression were treated with tamoxifen to induce the removal of the floxed STOP cassette, which prevented hACE2 expression. Before and after intranasal inoculation, the mice were weighed and clinically examined. On Days 5 and 10, the mice were sacrificed for isolation of internal organs and the further assessment of SARS-CoV-2 distribution. Intranasal SARS-CoV-2 inoculation in hACE2-LoxP(STOP)×UBC-Cre offspring resulted in weight loss and death in 6 out of 8 mice. Immunostaining and focus formation assays revealed the most significant viral load in the lung, brain, heart and intestine samples. In contrast, hACE2-LoxP(STOP) × Rosa-Cre offspring easily tolerated the infection, and SARS-CoV-2 was detected only in the brain and lungs, whereas other studied tissues had null or negligible levels of the virus. Histological examination revealed severe alterations in the lungs, and mild changes were observed in the brain tissues. Notably, no changes were observed in mice without tamoxifen treatment. Thus, this novel murine model with the Cre-dependent activation of hACE2 provides a useful and safe tool for COVID-19 studies.
Collapse
Affiliation(s)
- Alexander A. Dolskiy
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
- *Correspondence: Alexander A. Dolskiy,
| | - Andrey S. Gudymo
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Oleg S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Irina V. Grishchenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Ekaterina M. Shitik
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Dmitry Yu Prokopov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Vladislav O. Soldatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Laboratory of Genome Editing for Veterinary and Biomedicine, Belgorod State National Research University, Belgorod, Russia
| | - Elvira V. Sobolevskaya
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Sergey A. Bodnev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Natalia V. Danilchenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Anastasia A. Moiseeva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Polina Y. Torzhkova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Yulia A. Bulanovich
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Galina S. Onhonova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Elena K. Ivleva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Marina V. Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey E. Belykh
- Research Institute of General Pathology, Kursk State Medical University, Kursk, Russia
| | - Tatiana V. Tregubchak
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Alexander B. Ryzhikov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Elena V. Gavrilova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Rinat A. Maksyutov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| | - Alexey V. Deykin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Laboratory of Genome Editing for Veterinary and Biomedicine, Belgorod State National Research University, Belgorod, Russia
| | - Dmitry V. Yudkin
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, Koltsovo, Russia
| |
Collapse
|
248
|
Kim YI, Yu KM, Koh JY, Kim EH, Kim SM, Kim EJ, Casel MAB, Rollon R, Jang SG, Song MS, Park SJ, Jeong HW, Kim EG, Lee OJ, Kim YD, Choi Y, Lee SA, Choi YJ, Park SH, Jung JU, Choi YK. Age-dependent pathogenic characteristics of SARS-CoV-2 infection in ferrets. Nat Commun 2022; 13:21. [PMID: 35013229 PMCID: PMC8748994 DOI: 10.1038/s41467-021-27717-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
While the seroprevalence of SARS-CoV-2 in healthy people does not differ significantly among age groups, those aged 65 years or older exhibit strikingly higher COVID-19 mortality compared to younger individuals. To further understand differing COVID-19 manifestations in patients of different ages, three age groups of ferrets are infected with SARS-CoV-2. Although SARS-CoV-2 is isolated from all ferrets regardless of age, aged ferrets (≥3 years old) show higher viral loads, longer nasal virus shedding, and more severe lung inflammatory cell infiltration, and clinical symptoms compared to juvenile (≤6 months) and young adult (1–2 years) groups. Furthermore, direct contact ferrets co-housed with the virus-infected aged group shed more virus than direct-contact ferrets co-housed with virus-infected juvenile or young adult ferrets. Transcriptome analysis of aged ferret lungs reveals strong enrichment of gene sets related to type I interferon, activated T cells, and M1 macrophage responses, mimicking the gene expression profile of severe COVID-19 patients. Thus, SARS-CoV-2-infected aged ferrets highly recapitulate COVID-19 patients with severe symptoms and are useful for understanding age-associated infection, transmission, and pathogenesis of SARS-CoV-2. Here, Kim et al. characterize SARS-CoV-2 infection in juvenile, young, and old aged ferrets to provide a further understanding of differences in COVID-19 severity in humans at different ages. Aged ferrets have higher viral loads, shed virus longer, and mimic the transcriptomic profile of severely infected patients.
Collapse
Affiliation(s)
- Young-Il Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea.,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Kwang-Min Yu
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - June-Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eun-Ha Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Se-Mi Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea.,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Eun Ji Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Mark Anthony B Casel
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Rare Rollon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Seung-Gyu Jang
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Min-Suk Song
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Su-Jin Park
- Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| | - Hye Won Jeong
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Eung-Gook Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Ok-Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Yong-Dae Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Younho Choi
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shin-Ae Lee
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Youn Jung Choi
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jae U Jung
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Young Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea. .,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea. .,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.
| |
Collapse
|
249
|
Hwang YC, Lu RM, Su SC, Chiang PY, Ko SH, Ke FY, Liang KH, Hsieh TY, Wu HC. Monoclonal antibodies for COVID-19 therapy and SARS-CoV-2 detection. J Biomed Sci 2022; 29:1. [PMID: 34983527 PMCID: PMC8724751 DOI: 10.1186/s12929-021-00784-w] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is an exceptional public health crisis that demands the timely creation of new therapeutics and viral detection. Owing to their high specificity and reliability, monoclonal antibodies (mAbs) have emerged as powerful tools to treat and detect numerous diseases. Hence, many researchers have begun to urgently develop Ab-based kits for the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ab drugs for use as COVID-19 therapeutic agents. The detailed structure of the SARS-CoV-2 spike protein is known, and since this protein is key for viral infection, its receptor-binding domain (RBD) has become a major target for therapeutic Ab development. Because SARS-CoV-2 is an RNA virus with a high mutation rate, especially under the selective pressure of aggressively deployed prophylactic vaccines and neutralizing Abs, the use of Ab cocktails is expected to be an important strategy for effective COVID-19 treatment. Moreover, SARS-CoV-2 infection may stimulate an overactive immune response, resulting in a cytokine storm that drives severe disease progression. Abs to combat cytokine storms have also been under intense development as treatments for COVID-19. In addition to their use as drugs, Abs are currently being utilized in SARS-CoV-2 detection tests, including antigen and immunoglobulin tests. Such Ab-based detection tests are crucial surveillance tools that can be used to prevent the spread of COVID-19. Herein, we highlight some key points regarding mAb-based detection tests and treatments for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yu-Chyi Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Chieh Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Pao-Yin Chiang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Feng-Yi Ke
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Tzung-Yang Hsieh
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
250
|
Park JG, Pino PA, Akhter A, Alvarez X, Torrelles JB, Martinez-Sobrido L. Animal Models of COVID-19: Transgenic Mouse Model. Methods Mol Biol 2022; 2452:259-289. [PMID: 35554912 PMCID: PMC9563002 DOI: 10.1007/978-1-0716-2111-0_16] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), emerged in December 2019 in Wuhan, China, and rapidly spread throughout the world, threatening global public health. An animal model is a valuable and a crucial tool that allows understanding of nature in the pathogenesis of SARS-CoV-2 and its associated COVID-19 disease. Here we introduce detailed protocols of SARS-CoV-2 infection and COVID-19 disease using C57BL/6 (B6) transgenic mice expressing the human angiotensin-converting enzyme 2 (hACE2) from the human cytokeratin 18 promoter (K18 hACE2). To mimic natural SARS-CoV-2 infection, K18 hACE2 transgenic mice are infected intranasally under anesthesia. Upon infection, viral pathogenesis is determined by monitoring changes in body weight (morbidity) and monitoring survival (mortality), cytokine/chemokine responses, gross-lung pathology, histopathology, and viral replication in tissues. The presence of the virus and viral replication is evaluated by immunohistochemistry (IHC) and viral titrations, respectively, from the upper (nasal turbinate) and the lower (lungs) respiratory tracts, and nervous system (brain). Also, the immune response to SARS-CoV-2 infection is measured by cytokine/chemokine enzyme-linked immunosorbent assay (ELISA) from lung, spleen and brain homogenates to characterize the cytokine storm that hallmarks as one of the major causes of death caused by SARS-CoV-2 infection. This small rodent animal model based on the use of K18 hACE2 transgenic mice represents an excellent option to understand the pathogenicity of natural SARS-CoV-2 strains and its recently described Variants of Concern (VoC), and will be applicable to the identification and characterization of prophylactic (vaccine) and therapeutic (antiviral and/or neutralizing monoclonal antibodies) strategies for the prevention or treatment of SARS-CoV-2 infection or its associated COVID-19 disease.
Collapse
Affiliation(s)
- Jun-Gyu Park
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Paula A Pino
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Anwari Akhter
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Xavier Alvarez
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jordi B Torrelles
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Luis Martinez-Sobrido
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|