1
|
Awad NK. Organs on chips: fundamentals, bioengineering and applications. J Artif Organs 2025; 28:110-130. [PMID: 39134691 DOI: 10.1007/s10047-024-01460-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/12/2024] [Indexed: 05/16/2025]
Abstract
Human body constitutes unique biological system containing specific fluid mechanics and biomechanics. Traditional cell culture techniques of 2D and 3D do not recapitulate these specific natures of the human system. In addition, they lack the spatiotemporal conditions of representing the cells. Moreover, they do not enable the study of cell-cell interactions in multiple cell culture platforms. Therefore, establishing biological system of dynamic cell culture was of great interest. Organs on chips systems were fabricated proving their concept to mimic specific organs functions. Therefore, it paves the way for validating new drugs and establishes mechanisms of emerging diseases. It has played a key role in validating suitable vaccines for Coronavirus disease (COVID-19). Herein, the concept of organs on chips, fabrication methodology and their applications are discussed.
Collapse
Affiliation(s)
- Nasser K Awad
- Physical Chemistry Department, Advanced Materials Technology and Mineral Resources Research Institute, National Research Centre, Dokki, 12422, Cairo, Egypt.
| |
Collapse
|
2
|
Ishigaki H, Itoh Y. Translational research on pandemic virus infection using nonhuman primate models. Virology 2025; 606:110511. [PMID: 40139071 DOI: 10.1016/j.virol.2025.110511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
After the COVID-19 pandemic, nonhuman primate (NHP) models, which are necessary for the rapid development of vaccines and new medical therapies, have become important in studies on infectious diseases because of their genetic, metabolic, and immunological similarities to humans. Our group has long been using NHP models in studies on infectious diseases including H1N1 influenza pandemic and COVID-19. Despite limitations such as the limited number of animals and the husbandry requirements, NHP models have contributed to the prediction of the pathogenicity of emerging viruses and the evaluation of the efficacy of vaccines and therapeutics due to the similarity of NHP models to humans before starting clinical trials to select good candidates of vaccines and drugs. In this review, the findings obtained in NHP infectious disease models of influenza and COVID-19 are summarized to clarify the benefits of NHP models for studies on infectious diseases. We believe that this review will support future research in exploring new perspectives for the development of vaccines and therapies targeting influenza, COVID-19, and infectious diseases in future pandemics.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, 460 Setatsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, 460 Setatsukinowa, Otsu, Shiga, 520-2192, Japan; Central Research Laboratory, Shiga University of Medical Science, 205 Setatsukinowa, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
3
|
Dogadov DI, Kyuregyan KK, Minosyan AA, Goncharenko AM, Shmat EV, Mikhailov MI. [Acute respiratory viral infections in monkeys]. Vopr Virusol 2025; 70:7-24. [PMID: 40233333 DOI: 10.36233/0507-4088-293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Indexed: 04/17/2025]
Abstract
Acute respiratory viral infections (ARVI) are one of the most significant infections affecting the breeding of monkeys, especially among imported and captive primates. Respiratory diseases are also an important cause of morbidity and mortality in wild populations, and most of these infections can affect humans. Many anthropoid species, including apes, are susceptible to ARVI. Outbreaks of spontaneous respiratory infections have been described in many zoos and primatological centers around the world. Moreover, the study of spontaneous and experimental infection in laboratory primates provides an invaluable source of information on the biology and pathogenesis of ARVI and remains an indispensable tool for testing vaccines and drugs. The aim of this literature review was to summarize and analyze published data on the circulation of ARVI causative agents (parainfluenza viruses, adenoviruses, respiratory syncytial virus, influenza viruses, rhinoviruses, coronaviruses, metapneumoviruses, bocaviruses) among wild and captive primates, as well as the results of experimental modeling these infections in monkeys.
Collapse
Affiliation(s)
- D I Dogadov
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - K K Kyuregyan
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia
| | - A A Minosyan
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - A M Goncharenko
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - E V Shmat
- Sochi Institute (branch) of the Federal State Autonomous Educational Institution of Higher Education Peoples' Friendship University of Russia named after Patrice Lumumba
| | - M I Mikhailov
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| |
Collapse
|
4
|
Coish JM, MacNeil LA, MacNeil AJ. The SARS-CoV-2 antibody-dependent enhancement façade. Microbes Infect 2025; 27:105464. [PMID: 39662700 DOI: 10.1016/j.micinf.2024.105464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Antibody-dependent enhancement (ADE) is an immunological paradox whereby sensitization following a primary viral infection results in the subsequent enhancement of a similar secondary infection. This idiosyncratic immune response has been established in dengue virus infections, driven by four antigenically related serotypes co-circulating in endemic regions. Several coronaviruses exhibit antibody-mediated mechanisms of viral entry, which has led to speculation of an ADE capacity for SARS-CoV-2, though in vivo and epidemiological evidence do not currently support this phenomenon. Three distinct antibody-dependent mechanisms for SARS-CoV-2 entry have recently been demonstrated: 1. FcR-dependent, 2. ACE2-FcR-interdependent, and 3. FcR-independent. These mechanisms of viral entry may be dependent on SARS-CoV-2 antibody specificity; antibodies targeting the receptor binding domain (RBD) typically result in Fc-dependent and ACE2-FcR-interdependent entry, whereas antibodies targeting the N-terminal domain can induce a conformational change to the RBD that optimizes ACE2-receptor binding domain interactions independent of Fc receptors. Whether these antibody-dependent entry mechanisms of SARS-CoV-2 result in the generation of infectious progenies and enhancement of infection has not been robustly demonstrated. Furthermore, ADE of SARS-CoV-2 mediated by antigenic seniority remains a theoretical concern, as no evidence suggests that SARS-CoV-2 imprinting blunts a subsequent immune response, contributing to severe COVID-19 disease.
Collapse
Affiliation(s)
- Jeremia M Coish
- Department of Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Lori A MacNeil
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada.
| |
Collapse
|
5
|
Vanderheiden A, Diamond MS. Animal Models of Non-Respiratory, Post-Acute Sequelae of COVID-19. Viruses 2025; 17:98. [PMID: 39861887 PMCID: PMC11768974 DOI: 10.3390/v17010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Post-acute sequelae of COVID-19 (PASC) are a diverse set of symptoms and syndromes driven by dysfunction of multiple organ systems that can persist for years and negatively impact the quality of life for millions of individuals. We currently lack specific therapeutics for patients with PASC, due in part to an incomplete understanding of its pathogenesis, especially for non-pulmonary sequelae. Here, we discuss three animal models that have been utilized to investigate PASC: non-human primates (NHPs), hamsters, and mice. We focus on neurological, gastrointestinal, and cardiovascular PASC and highlight advances in mechanistic insight that have been made using these animal models, as well as discussing the sequelae that warrant continued and intensive research.
Collapse
Affiliation(s)
- Abigail Vanderheiden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
6
|
Kimotho J, Sein Y, Sayed S, Shah R, Mwai K, Saleh M, Wanjiku P, Mwacharo J, Nyagwange J, Karanja H, Kutima B, Gitonga JN, Mugo D, Karanu A, Moranga L, Oluoch V, Shah J, Mutiso J, Mburu A, Nneka Z, Betti P, Usyu Mutinda W, Issak Abdi A, Bejon P, Isabella Ochola-Oyier L, M.Warimwe G, Nduati EW, M. Ndungu F. Kinetics of naturally induced binding and neutralising anti-SARS-CoV-2 antibody levels and potencies among SARS-CoV-2 infected Kenyans with diverse grades of COVID-19 severity: an observational study. Wellcome Open Res 2024; 8:350. [PMID: 39640868 PMCID: PMC11617823 DOI: 10.12688/wellcomeopenres.19414.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background Given the low levels of coronavirus disease 2019 (COVID-19) vaccine coverage in sub-Saharan Africa (sSA), despite high levels of natural severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) exposures, strategies for extending the breadth and longevity of naturally acquired immunity are warranted. Designing such strategies will require a good understanding of naturally acquired immunity. Methods We measured whole-spike immunoglobulin G (IgG) and spike-receptor binding domain (RBD) total immunoglobulins (Igs) on 585 plasma samples collected longitudinally over five successive time points within six months of COVID-19 diagnosis in 309 COVID-19 patients. We measured antibody-neutralising potency against the wild-type (Wuhan) SARS-CoV-2 pseudovirus in a subset of 51 patients over three successive time points. Binding and neutralising antibody levels and potencies were then tested for correlations with COVID-19 severities. Results Rates of seroconversion increased from day 0 (day of PCR testing) to day 180 (six months) (63.6% to 100 %) and (69.3 % to 97%) for anti-spike-IgG and anti-spike-RBD binding Igs, respectively. Levels of these binding antibodies peaked at day 28 (p<0.01) and were subsequently maintained for six months without significant decay (p>0.99). Similarly, antibody-neutralising potencies peaked at day 28 (p<0.01) but declined by three-fold, six months after COVID-19 diagnosis (p<0.01). Binding antibody levels were highly correlated with neutralising antibody potencies at all the time points analysed (r>0.60, p<0.01). Levels and potencies of binding and neutralising antibodies increased with disease severity. Conclusions Most COVID-19 patients generated SARS-CoV-2 specific binding antibodies that remained stable in the first six months of infection. However, the respective neutralising antibodies decayed three-fold by month-six of COVID-19 diagnosis suggesting that they are short-lived, consistent with what has been observed elsewhere in the world. Thus, regular vaccination boosters are required to sustain the high levels of anti-SARS-CoV-2 naturally acquired neutralising antibody potencies in our population.
Collapse
Affiliation(s)
- John Kimotho
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
| | - Yiakon Sein
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Shahin Sayed
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Reena Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Kennedy Mwai
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Mansoor Saleh
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Perpetual Wanjiku
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Jedidah Mwacharo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - James Nyagwange
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Henry Karanja
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Bernadette Kutima
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - John N. Gitonga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Ann Karanu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Linda Moranga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Viviane Oluoch
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Jasmit Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Julius Mutiso
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Alfred Mburu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Zaitun Nneka
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Peter Betti
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | | | - Abdirahman Issak Abdi
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - George M.Warimwe
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eunice W. Nduati
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Francis M. Ndungu
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Byrne C, Schiffer JT. Ensemble modeling of SARS-CoV-2 immune dynamics in immunologically naïve rhesus macaques predicts that potent, early innate immune responses drive viral elimination. Front Immunol 2024; 15:1426016. [PMID: 39575237 PMCID: PMC11578959 DOI: 10.3389/fimmu.2024.1426016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction An unprecedented breadth of longitudinal viral and multi-scale immunological data has been gathered during SARS-CoV-2 infection. However, due to the high complexity, non-linearity, multi-dimensionality, mixed anatomic sampling, and possible autocorrelation of available immune data, it is challenging to identify the components of the innate and adaptive immune response that drive viral elimination. Novel mathematical models and analytical approaches are required to synthesize contemporaneously gathered cytokine, transcriptomic, flow cytometry, antibody response, and viral load data into a coherent story of viral control, and ultimately to discriminate drivers of mild versus severe infection. Methods We investigated a dataset describing innate, SARS-CoV-2 specific T cell, and antibody responses in the lung during early and late stages of infection in immunologically naïve rhesus macaques. We used multi-model inference and ensemble modeling approaches from ecology and weather forecasting to compare and combine various competing models. Results and discussion Model outputs suggest that the innate immune response plays a crucial role in controlling early infection, while SARS-CoV-2 specific CD4+ T cells correspond to later viral elimination, and anti-spike IgG antibodies do not impact viral dynamics. Among the numerous genes potentially contributing to the innate response, we identified IFI27 as most closely linked to viral load decline. A 90% knockdown of the innate response from our validated model resulted in a ~10-fold increase in peak viral load during infection. Our approach provides a novel methodological framework for future analyses of similar complex, non-linear multi-component immunologic data sets.
Collapse
Affiliation(s)
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center,
Seattle, WA, United States
| |
Collapse
|
8
|
Wang C, Liu S, Li C, Wang Z, Ming R, Huang L. Monitoring the Cascade of Monocyte-Derived Macrophages to Influenza Virus Infection in Human Alveolus Chips. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60045-60055. [PMID: 39450775 DOI: 10.1021/acsami.4c15125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Respiratory viruses ravage the world and seriously threaten people's health. Despite intense research efforts, the immune mechanism underlying respiratory virus-induced acute lung injury (ALI) and pulmonary fibrosis (PF) has not been fully elucidated. Here, the cascade of monocyte-derived macrophages to influenza A virus infection is monitored on an optimized human alveolus chip to reveal the role of macrophages in the development of ALI and PF. We find that viral infection causes damage to the alveolar air-liquid barrier and the release of inflammatory cytokines, which induce the M0 macrophages to gather and polarize to the M1 phenotype at the damaged site through recruitment, adhesion, migration, and activation, leading to ALI. Afterward, M1 macrophages polarize into the M2 phenotype, and then transform into myofibroblasts, followed by enhanced secretion of various anti-inflammatory cytokines and profibrotic cytokines, to promote PF. Our study provides an insight into the pathogenesis of virus-induced ALI and PF, which will assist in the development of therapeutic strategies and drugs for treating influenza and other respiratory virus infections.
Collapse
Affiliation(s)
- Chenguang Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Shujun Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chuyu Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhongjie Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Ruiqi Ming
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lili Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
- Tangshan Research Institute, Beijing Institute of Technology, Tangshan 063000, P. R. China
| |
Collapse
|
9
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
10
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
11
|
Luo Y, Zhang Z, Ren J, Dou C, Wen J, Yang Y, Li X, Yan Z, Han Y. SARS-Cov-2 spike induces intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. Front Immunol 2024; 15:1303356. [PMID: 38686388 PMCID: PMC11056506 DOI: 10.3389/fimmu.2024.1303356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Background Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), as a typical tumor marker, has been found to exert immunomodulatory effects in many diseases. We previously reported the clinical and molecular evidences supporting that SARS-Cov-2 infected the gastrointestinal (GI) tract and found a reduction of CEACAM5 in COVID-19 patients' feces which associated with gut dysbiosis. Yet the role of CEACAM5 in GI infection is ill-defined. Methods Mice models were established through intraperitoneally injecting with recombinant viral spike-Fc to mimic the intestinal inflammation. We collected duodenum, jejunum, ileum and colon samples after 6h, 2 days, 4 days and 7 days of spike-Fc or control-Fc injection to perform proteomic analysis. Blood was collected from healthy donors and peripheral blood mononuclear cells (PBMC) were separated by density gradient centrifugation, then CD4+ T cells were isolated with magnetic beads and co-cultured with Caco-2 cells. Results In addition to intestinal CEACAM5, the expression of tight junction and the percent of CD4+ T lymphocytes were significantly decreased in spike-Fc group compared to control (p < 0.05), accompanied with increased level of inflammatory factors. The KEGG analysis revealed differentially expressed proteins were mainly enriched in the coronavirus disease (COVID-19), tight junction, focal adhesion, adherens junction and PI3K-Akt signaling pathway. Protein-protein interaction (PPI) network analysis identified the interaction between CEACAM5 and Galectin-9 that was also verified by molecular docking and co-IP assay. We further confirmed a reduction of CEACAM5 in SARS-CoV-2 spike stimulated enterocytes could promote the expression of Galectin-9 protein in CD4+T cells. Then it gave rise to the increasing release of inflammatory factors and increased apoptosis of CD4+T cells by inhibition of PI3K/AKT/mTOR pathway. Ultimately intestinal barrier dysfunction happened. Conclusion Our results indicated that CEACAM5 overexpression and Galectin-9 knockdown played a protective role in intestinal barrier injury upon spike-Fc stimulation. Collectively, our findings identified firstly that SARS-CoV-2 spike induced intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. The result provides potential therapeutic targets in intestinal barrier dysfunction for treating severe COVID patients.
Collapse
Affiliation(s)
- Yingshu Luo
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhenling Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiangnan Ren
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Chunxu Dou
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiancheng Wen
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yang Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhixiang Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yanzhi Han
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| |
Collapse
|
12
|
Deng W, Bao L, Song Z, Zhang L, Yu P, Xu Y, Wang J, Zhao W, Zhang X, Han Y, Li Y, Liu J, Lv Q, Liang X, Li F, Qi F, Deng R, Wang S, Xiong Y, Xiao R, Wang H, Qin C. Infection with SARS-CoV-2 can cause pancreatic impairment. Signal Transduct Target Ther 2024; 9:98. [PMID: 38609366 PMCID: PMC11014980 DOI: 10.1038/s41392-024-01796-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/25/2024] [Accepted: 03/06/2024] [Indexed: 04/14/2024] Open
Abstract
Evidence suggests associations between COVID-19 patients or vaccines and glycometabolic dysfunction and an even higher risk of the occurrence of diabetes. Herein, we retrospectively analyzed pancreatic lesions in autopsy tissues from 67 SARS-CoV-2 infected non-human primates (NHPs) models and 121 vaccinated and infected NHPs from 2020 to 2023 and COVID-19 patients. Multi-label immunofluorescence revealed direct infection of both exocrine and endocrine pancreatic cells by the virus in NHPs and humans. Minor and limited phenotypic and histopathological changes were observed in adult models. Systemic proteomics and metabolomics results indicated metabolic disorders, mainly enriched in insulin resistance pathways, in infected adult NHPs, along with elevated fasting C-peptide and C-peptide/glucose ratio levels. Furthermore, in elder COVID-19 NHPs, SARS-CoV-2 infection causes loss of beta (β) cells and lower expressed-insulin in situ characterized by islet amyloidosis and necrosis, activation of α-SMA and aggravated fibrosis consisting of lower collagen in serum, an increase of pancreatic inflammation and stress markers, ICAM-1 and G3BP1, along with more severe glycometabolic dysfunction. In contrast, vaccination maintained glucose homeostasis by activating insulin receptor α and insulin receptor β. Overall, the cumulative risk of diabetes post-COVID-19 is closely tied to age, suggesting more attention should be paid to blood sugar management in elderly COVID-19 patients.
Collapse
Affiliation(s)
- Wei Deng
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Linlin Bao
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Zhiqi Song
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Ling Zhang
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Pin Yu
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Yanfeng Xu
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Jue Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Wenjie Zhao
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Yunlin Han
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Yanhong Li
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Jiangning Liu
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Qi Lv
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Xujian Liang
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Fengdi Li
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Feifei Qi
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Ran Deng
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Siyuan Wang
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Yibai Xiong
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Ruiping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
| | - Hongyang Wang
- Chinese Academy of Engineering, Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Yangpu District, Shanghai, 200438, China.
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, PR China.
- National Laboratory for Oncogenes and Related Genes, Cancer Institute of Shanghai Jiao Tong University, Shanghai, 200441, PR China.
| | - Chuan Qin
- NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China.
- Changping National laboratory (CPNL), Beijing, 102206, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, National Health Commission of the People's Republic of China, Beijing, PR China.
| |
Collapse
|
13
|
Chen Z, Yuan Y, Hu Q, Zhu A, Chen F, Li S, Guan X, Lv C, Tang T, He Y, Cheng J, Zheng J, Hu X, Zhao J, Zhao J, Sun J. SARS-CoV-2 immunity in animal models. Cell Mol Immunol 2024; 21:119-133. [PMID: 38238440 PMCID: PMC10806257 DOI: 10.1038/s41423-023-01122-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.
Collapse
Affiliation(s)
- Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yaochang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Shu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xin Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Chao Lv
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xiaoyu Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518005, China.
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
14
|
El-Baky NA, Amara AA, Uversky VN, Redwan EM. Intrinsic factors behind long COVID: III. Persistence of SARS-CoV-2 and its components. J Cell Biochem 2024; 125:22-44. [PMID: 38098317 DOI: 10.1002/jcb.30514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 01/16/2024]
Abstract
Considerable research has been done in investigating SARS-CoV-2 infection, its characteristics, and host immune response. However, debate is still ongoing over the emergence of post-acute sequelae of SARS-CoV-2 infection (PASC). A multitude of long-lasting symptoms have been reported several weeks after the primary acute SARS-CoV-2 infection that resemble several other viral infections. Thousands of research articles have described various post-COVID-19 conditions. Yet, the evidence around these ongoing health problems, the reasons behind them, and their molecular underpinnings are scarce. These persistent symptoms are also known as long COVID-19. The persistence of SARS-CoV-2 and/or its components in host tissues can lead to long COVID. For example, the presence of viral nucleocapsid protein and RNA was detected in the skin, appendix, and breast tissues of some long COVID patients. The persistence of viral RNA was reported in multiple anatomic sites, including non-respiratory tissues such as the adrenal gland, ocular tissue, small intestine, lymph nodes, myocardium, and sciatic nerve. Distinctive viral spike sequence variants were also found in non-respiratory tissues. Interestingly, prolonged detection of viral subgenomic RNA was observed across all tissues, sometimes in multiple tissues of the same patient, which likely reflects recent but defective viral replication. Moreover, the persistence of SARS-CoV-2 RNA was noticed throughout the brain at autopsy, as late as 230 days following symptom onset among unvaccinated patients who died of severe infection. Here, we review the persistence of SARS-CoV-2 and its components as an intrinsic factor behind long COVID. We also highlight the immunological consequences of this viral persistence.
Collapse
Affiliation(s)
- Nawal Abd El-Baky
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Egypt
| | - Amro A Amara
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Elrashdy M Redwan
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
McKendry R, Lemm NM, Papargyris L, Chiu C. Human Challenge Studies with Coronaviruses Old and New. Curr Top Microbiol Immunol 2024; 445:69-108. [PMID: 35181805 DOI: 10.1007/82_2021_247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus infections have been known to cause disease in animals since as early as the 1920s. However, only seven coronaviruses capable of causing human disease have been identified thus far. These Human Coronaviruses (HCoVs) include the causes of the common cold, but more recent coronaviruses that have emerged (i.e. SARS-CoV, MERS-CoV and SARS-CoV-2) are associated with much greater morbidity and mortality. HCoVs have been relatively under-studied compared to other common respiratory infections, as historically they have presented with mild symptoms. This has led to a relatively limited understanding of their animal reservoirs, transmission and determinants of immune protection. To address this, human infection challenge studies with HCoVs have been performed that enable a detailed clinical and immunological analysis of the host response at specific time points under controlled conditions with standardised viral inocula. Until recently, all such human challenge studies were conducted with common cold HCoVs, with the study of SARS-CoV and MERS-CoV unacceptable due to their greater pathogenicity. However, with the emergence of SARS-CoV-2 and the COVID-19 pandemic during which severe outcomes in young healthy adults have been rare, human challenge studies with SARS-CoV-2 are now being developed. Two SARS-CoV-2 human challenge studies in the UK studying individuals with and without pre-existing immunity are underway. As well as providing a platform for testing of antivirals and vaccines, such studies will be critical for understanding the factors associated with susceptibility to SARS-CoV-2 infection and thus developing improved strategies to tackle the current as well as future HCoV pandemics. Here, we summarise the major questions about protection and pathogenesis in HCoV infection that human infection challenge studies have attempted to answer historically, as well as the knowledge gaps that aim to be addressed with contemporary models.
Collapse
Affiliation(s)
- Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
16
|
Diego JGB, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian hamsters. PLoS Pathog 2024; 20:e1011805. [PMID: 38198521 PMCID: PMC10805310 DOI: 10.1371/journal.ppat.1011805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 01/23/2024] [Accepted: 11/06/2023] [Indexed: 01/12/2024] Open
Abstract
Hybrid immunity (vaccination + natural infection) to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. The mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed for T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera, reflected by smaller antigenic cartography distances. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kim Handrejk
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sara S. El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Max W. Chang
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Brett T. Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, United States of America
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
17
|
Termansen MB, Frische S. Fecal-oral transmission of SARS-CoV-2: A systematic review of evidence from epidemiological and experimental studies. Am J Infect Control 2023; 51:1430-1437. [PMID: 37121473 PMCID: PMC10141930 DOI: 10.1016/j.ajic.2023.04.170] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND SARS-CoV-2 ribonucleic acid (RNA) has been detected in feces, but RNA is not infectious. This systematic review aims to answer if fecal SARS-CoV-2 is experimentally infectious and if evidence of human fecal-oral SARS-CoV-2 transmission exists. METHODS On September 19, 2022, we searched PubMed, Embase, Web of Science, medRxiv, and bioRxiv. Biomedical studies inoculating SARS-CoV-2 from feces, rectal, or anal swabs in cells, tissue, organoids, or animals were included. Epidemiological studies of groups differing in exposure to fecal SARS-CoV-2 were included. Risk of bias was assessed using standardized tools. Results were summarized by vote counting, tabulation, and a harvest plot. PROSPERO registration no. CRD42020221719. RESULTS A total of 4,874 studies were screened; 26 studies were included; and 13 out of 23 biomedical studies (56.5%) succeeded in infection. Two (66.7%) epidemiological studies found limited evidence suggesting fecal-oral transmission. All studies had concerns about the risk of bias. CONCLUSIONS It is possible to experimentally infect cell cultures, organoids, and animals with fecal SARS-CoV-2. No strong epidemiologic evidence was found to support human fecal-oral transmission. We advise future research to study fecal infectivity at different time points during infection, apply appropriate controls, use in vivo models, and study fecal exposure as a risk factor of transmission in human populations.
Collapse
|
18
|
Rong N, Liu J. Development of animal models for emerging infectious diseases by breaking the barrier of species susceptibility to human pathogens. Emerg Microbes Infect 2023; 12:2178242. [PMID: 36748729 PMCID: PMC9970229 DOI: 10.1080/22221751.2023.2178242] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Outbreaks of emerging infectious diseases pose a serious threat to public health security, human health and economic development. After an outbreak, an animal model for an emerging infectious disease is urgently needed for studying the etiology, host immune mechanisms and pathology of the disease, evaluating the efficiency of vaccines or drugs against infection, and minimizing the time available for animal model development, which is usually hindered by the nonsusceptibility of common laboratory animals to human pathogens. Thus, we summarize the technologies and methods that induce animal susceptibility to human pathogens, which include viral receptor humanization, pathogen-targeted tissue humanization, immunodeficiency induction and screening for naturally susceptible animal species. Furthermore, the advantages and deficiencies of animal models developed using each method were analyzed, and these will guide the selection of susceptible animals and potentially reduce the time needed to develop animal models during epidemics.
Collapse
Affiliation(s)
- Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, People’s Republic of China, Jiangning Liu
| |
Collapse
|
19
|
Mabry ME, Fanelli A, Mavian C, Lorusso A, Manes C, Soltis PS, Capua I. The panzootic potential of SARS-CoV-2. Bioscience 2023; 73:814-829. [PMID: 38125826 PMCID: PMC10728779 DOI: 10.1093/biosci/biad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/09/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023] Open
Abstract
Each year, SARS-CoV-2 is infecting an increasingly unprecedented number of species. In the present article, we combine mammalian phylogeny with the genetic characteristics of isolates found in mammals to elaborate on the host-range potential of SARS-CoV-2. Infections in nonhuman mammals mirror those of contemporary viral strains circulating in humans, although, in certain species, extensive viral circulation has led to unique genetic signatures. As in other recent studies, we found that the conservation of the ACE2 receptor cannot be considered the sole major determinant of susceptibility. However, we are able to identify major clades and families as candidates for increased surveillance. On the basis of our findings, we argue that the use of the term panzootic could be a more appropriate term than pandemic to describe the ongoing scenario. This term better captures the magnitude of the SARS-CoV-2 host range and would hopefully inspire inclusive policy actions, including systematic screenings, that could better support the management of this worldwide event.
Collapse
Affiliation(s)
- Makenzie E Mabry
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, United States
| | - Angela Fanelli
- Department of Veterinary Medicine, University of Bari, Valenzano, Bari, Italy
| | - Carla Mavian
- Emerging Pathogens Institute and with the Department of Pathology, University of Florida, Gainesville, Florida, United States
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise G. Caporale, Teramo, Italy
| | - Costanza Manes
- Department of Wildlife Ecology and Conservation and with the One Health Center of Excellence, University of Florida, Gainesville, Florida, United States
| | - Pamela S Soltis
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, United States
| | - Ilaria Capua
- One Health Center of Excellence, University of Florida, Gainesville, Florida, United States
- School of International Advanced Studies, Johns Hopkins University, Bologna, Italy
| |
Collapse
|
20
|
Li J, Wang J, Wang H. Emerging Landscape of Preclinical Models for Studying COVID-19 Neurologic Diseases. ACS Pharmacol Transl Sci 2023; 6:1323-1339. [PMID: 37854617 PMCID: PMC10580392 DOI: 10.1021/acsptsci.3c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 10/20/2023]
Abstract
COVID-19 (Coronavirus Disease 2019) is an infectious disease caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) and has globally infected 768 million people and caused over 6 million deaths. COVID-19 primarily affects the respiratory system but increasing reports of neurologic symptoms associated with COVID-19 have been reported in the literature. The exact mechanism behind COVID-19 neurologic pathophysiology remains poorly understood due to difficulty quantifying clinical neurologic symptoms in humans and correlating them to findings in human post-mortem samples and animal models. Thus, robust preclinical experimental models for COVID-19 neurologic manifestations are urgently needed. Here, we review recent advances in in vitro, in vivo, and other models and technologies for studying COVID-19 including primary cell cultures, pluripotent stem cell-derived neurons and organoids, rodents, nonhuman primates, 3D bioprinting, artificial intelligence, and multiomics. We specifically focus our discussion on the contribution, recent advancements, and limitations these preclinical models have on furthering our understanding of COVID-19's neuropathic physiology. We also discuss these models' roles in the screening and development of therapeutics, vaccines, antiviral drugs, and herbal medicine, and on future opportunities for COVID-19 neurologic research and clinical management.
Collapse
Affiliation(s)
- Jason Li
- Department
of Neurology, Indiana University School
of Medicine, Indianapolis, Indiana 46202, United States
| | - Jing Wang
- Department
of Cellular and Molecular Medicine, University
of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Hu Wang
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore 21215, United States
| |
Collapse
|
21
|
Francis ME, Jansen EB, Yourkowski A, Selim A, Swan CL, MacPhee BK, Thivierge B, Buchanan R, Lavender KJ, Darbellay J, Rogers MB, Lew J, Gerdts V, Falzarano D, Skowronski DM, Sjaarda C, Kelvin AA. Previous infection with seasonal coronaviruses does not protect male Syrian hamsters from challenge with SARS-CoV-2. Nat Commun 2023; 14:5990. [PMID: 37752151 PMCID: PMC10522707 DOI: 10.1038/s41467-023-41761-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
SARS-CoV-2 variants and seasonal coronaviruses continue to cause disease and coronaviruses in the animal reservoir pose a constant spillover threat. Importantly, understanding of how previous infection may influence future exposures, especially in the context of seasonal coronaviruses and SARS-CoV-2 variants, is still limited. Here we adopted a step-wise experimental approach to examine the primary immune response and subsequent immune recall toward antigenically distinct coronaviruses using male Syrian hamsters. Hamsters were initially inoculated with seasonal coronaviruses (HCoV-NL63, HCoV-229E, or HCoV-OC43), or SARS-CoV-2 pango B lineage virus, then challenged with SARS-CoV-2 pango B lineage virus, or SARS-CoV-2 variants Beta or Omicron. Although infection with seasonal coronaviruses offered little protection against SARS-CoV-2 challenge, HCoV-NL63-infected animals had an increase of the previously elicited HCoV-NL63-specific neutralizing antibodies during challenge with SARS-CoV-2. On the other hand, primary infection with HCoV-OC43 induced distinct T cell gene signatures. Gene expression profiling indicated interferon responses and germinal center reactions to be induced during more similar primary infection-challenge combinations while signatures of increased inflammation as well as suppression of the antiviral response were observed following antigenically distant viral challenges. This work characterizes and analyzes seasonal coronaviruses effect on SARS-CoV-2 secondary infection and the findings are important for pan-coronavirus vaccine design.
Collapse
Affiliation(s)
- Magen E Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ethan B Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alaa Selim
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cynthia L Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brian K MacPhee
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brittany Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kerry J Lavender
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Matthew B Rogers
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyne Lew
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Danuta M Skowronski
- BC Centre for Disease Control, Immunization Programs and Vaccine Preventable Diseases Service, Vancouver, BC, Canada
- University of British Columbia, School of Population and Public Health, Vancouver, BC, Canada
| | - Calvin Sjaarda
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
- Queen's Genomics Lab at Ongwanada (Q-GLO), Ongwanada Resource Centre, Kingston, ON, Canada
| | - Alyson A Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
22
|
Zhang Y, Li Q, Liu N, Hu J, Lin X, Huang M, Wei Y, Qi X, Chen X. Secure reversal of immune evasion from refractory NSCLC and highly contagious CoV-2 mutants by using 3D-engineered multifunctional biologics. Bioeng Transl Med 2023; 8:e10554. [PMID: 37693048 PMCID: PMC10487317 DOI: 10.1002/btm2.10554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/22/2023] [Accepted: 05/16/2023] [Indexed: 09/12/2023] Open
Abstract
There is an imperative choice to develop a secure feasible strategy to address evasion dynamics of refractory tumors and SARS-CoV-2-variants, while stem cell-based protocol may be more reliable as its unique ability for resetting multifunctional immunity to address progressive tumor and the constantly-evolving virus. In this study, spheroid-embryonoid stem cells from mature somatic cells were engineered as multifunctional biologics (3D-E/BSC) and inoculated in senile rhesus to identify secure potential against immune-evasion from viral-variants. Meanwhile, a cohort of eligible patients with stage IV NSCLC were approved for phase I clinical trials. Subsequently, long-lasting security and efficacy were validated by primate and clinical trials (p < 0.01) in that it could not only stimulate serological immunity, but also reset core immunity for hosts to address variant evasion after 3D-E/BSC withdrawal. Particularly, illustrated by single-cell evolving trajectory, 3D-E/BSC had securely reset senile thymus of aging hosts to remodel core immunity by rearranging naive rhythm to evolve TRGC2+/JCHAIN+NKT clusters to abolish tumoral and viral evasion dynamics with path-feedbacks of NSCLC and COVID-19 simultaneously activated, leading to continuous blockade of breakthrough infection of viral-mutants and long-term survival in one-third of terminal patients without adjuvant required. Our study may pioneer a practical multifunctional strategy to eliminate evasion of SARS-CoV-2 variants and refractory NSCLC so as for victims to restart a new life-equation.
Collapse
Affiliation(s)
- Yanna Zhang
- Department of Blood Transfusion, Sichuan Provincial People’s HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Qian Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Nanxi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jianchuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xiaojuan Lin
- Department of Gynecology & Obstetrics, West China Second HospitalSichuan UniversityChengduChina
| | - Meijuan Huang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Yuquan Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Xiaorong Qi
- Department of Gynecology & Obstetrics, West China Second HospitalSichuan UniversityChengduChina
| | - Xiancheng Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
23
|
Coelho GM, Cataneo AHD, Raboni SM, Nogueira MB, de Paula CBV, Almeida ACSF, Rogerio VZ, Zanchin NT, de Noronha L, Zanluca C, Duarte Dos Santos CN. Development of an anti-SARS-CoV-2 monoclonal antibody panel and its applicability as a reagent in high-throughput fluorescence reduction neutralization and immunohistochemistry assays. J Med Virol 2023; 95:e29111. [PMID: 37750235 DOI: 10.1002/jmv.29111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/01/2023] [Accepted: 09/10/2023] [Indexed: 09/27/2023]
Abstract
Since its emergence in late 2019, coronavirus disease 2019 (COVID-19) has caused millions of deaths and socioeconomic losses. Although vaccination significantly reduced disease mortality, it has been shown that protection wanes over time, and that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) may escape vaccine-derived immunity. Therefore, serological studies are necessary to assess protection in the population and guide vaccine regimens. A common measure of protective immunity is the presence of neutralizing antibodies (nAbs). However, the gold standard for measuring nAbs (plaque reduction neutralization test, or PRNT) is laborious and time-consuming, limiting its large-scale applicability. We developed a high-throughput fluorescence reduction neutralization assay (FRNA) to detect SARS-CoV-2 nAbs. Because the assay relies on immunostaining, we developed and characterized monoclonal antibodies (mAbs) to lower costs and reduce the assay's vulnerability to reagent shortages. Using samples of individuals vaccinated with COVID-19 and unvaccinated/pre-pandemic samples, we showed that FRNA results using commercial and in-house mAbs strongly correlated with those of the PRNT method while providing results in 70% less time. In addition to providing a fast, reliable, and high-throughput alternative for measuring nAbs, the FRNA can be easily customized to assess SARS-CoV-2 VOCs. Additionally, the mAb we produced was able to detect SARS-CoV-2 in pulmonary tissues by immunohistochemistry assays.
Collapse
Affiliation(s)
- Gabriela M Coelho
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/Fiocruz), Curitiba, Paraná, Brasil
| | - Allan H D Cataneo
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/Fiocruz), Curitiba, Paraná, Brasil
| | - Sonia M Raboni
- Complexo Hospital de Clínicas, Universidade Federal Do Paraná, Curitiba, Paraná, Brasil
| | - Meri B Nogueira
- Complexo Hospital de Clínicas, Universidade Federal Do Paraná, Curitiba, Paraná, Brasil
| | - Caroline B Vaz de Paula
- Laboratório de Patologia Experimental, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brasil
| | - Ana C S F Almeida
- Laboratório de Patologia Experimental, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brasil
| | - Vanessa Z Rogerio
- Laboratório de Biologia Estrutural e Engenharia de Proteínas, Instituto Carlos Chagas (ICC/Fiocruz), Curitiba, Paraná, Brasil
| | - Nilson T Zanchin
- Laboratório de Biologia Estrutural e Engenharia de Proteínas, Instituto Carlos Chagas (ICC/Fiocruz), Curitiba, Paraná, Brasil
| | - Lucia de Noronha
- Laboratório de Patologia Experimental, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brasil
| | - Camila Zanluca
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/Fiocruz), Curitiba, Paraná, Brasil
| | | |
Collapse
|
24
|
Sha A, Liu Y, Hao H. Current state-of-the-art and potential future therapeutic drugs against COVID-19. Front Cell Dev Biol 2023; 11:1238027. [PMID: 37691829 PMCID: PMC10485263 DOI: 10.3389/fcell.2023.1238027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
The novel coronavirus disease (COVID-19) continues to endanger human health, and its therapeutic drugs are under intensive research and development. Identifying the efficacy and toxicity of drugs in animal models is helpful for further screening of effective medications, which is also a prerequisite for drugs to enter clinical trials. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) invades host cells mainly by the S protein on its surface. After the SARS-CoV-2 RNA genome is injected into the cells, M protein will help assemble and release new viruses. RdRp is crucial for virus replication, assembly, and release of new virus particles. This review analyzes and discusses 26 anti-SARS-CoV-2 drugs based on their mechanism of action, effectiveness and safety in different animal models. We propose five drugs to be the most promising to enter the next stage of clinical trial research, thus providing a reference for future drug development.
Collapse
Affiliation(s)
- Ailong Sha
- School of Teacher Education, Chongqing Three Gorges University, Chongqing, China
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Yi Liu
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Haiyan Hao
- School of Environmental and Chemical Engineering, Chongqing, China
| |
Collapse
|
25
|
Corleis B, Bastian M, Hoffmann D, Beer M, Dorhoi A. Animal models for COVID-19 and tuberculosis. Front Immunol 2023; 14:1223260. [PMID: 37638020 PMCID: PMC10451089 DOI: 10.3389/fimmu.2023.1223260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Respiratory infections cause tremendous morbidity and mortality worldwide. Amongst these diseases, tuberculosis (TB), a bacterial illness caused by Mycobacterium tuberculosis which often affects the lung, and coronavirus disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2), stand out as major drivers of epidemics of global concern. Despite their unrelated etiology and distinct pathology, these infections affect the same vital organ and share immunopathogenesis traits and an imperative demand to model the diseases at their various progression stages and localizations. Due to the clinical spectrum and heterogeneity of both diseases experimental infections were pursued in a variety of animal models. We summarize mammalian models employed in TB and COVID-19 experimental investigations, highlighting the diversity of rodent models and species peculiarities for each infection. We discuss the utility of non-human primates for translational research and emphasize on the benefits of non-conventional experimental models such as livestock. We epitomize advances facilitated by animal models with regard to understanding disease pathophysiology and immune responses. Finally, we highlight research areas necessitating optimized models and advocate that research of pulmonary infectious diseases could benefit from cross-fertilization between studies of apparently unrelated diseases, such as TB and COVID-19.
Collapse
Affiliation(s)
- Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Max Bastian
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
26
|
Li Z, Xiang T, Liang B, Liu J, Deng H, Yang X, Wang H, Feng X, Zelinskyy G, Trilling M, Sutter K, Lu M, Dittmer U, Wang B, Yang D, Zheng X, Liu J. SARS-CoV-2-specific T cell responses wane profoundly in convalescent individuals 10 months after primary infection. Virol Sin 2023; 38:606-619. [PMID: 37414153 PMCID: PMC10436107 DOI: 10.1016/j.virs.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
A key question in the coronavirus disease 2019 (COVID-19) pandemic is the duration of specific T cell responses against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) post primary infection, which is difficult to address due to the large-scale COVID-19 vaccination and re-exposure to the virus. Here, we conducted an analysis of the long-term SARS-CoV-2-specific T cell responses in a unique cohort of convalescent individuals (CIs) that were among the first to be infected worldwide and without any possible antigen re-exposure since then. The magnitude and breadth of SARS-CoV-2-specific T cell responses correlated inversely with the time that had elapsed from disease onset and the age of those CIs. The mean magnitude of SARS-CoV-2-specific CD4 and CD8 T cell responses decreased about 82% and 76%, respectively, over the time period of ten months after infection. Accordingly, the longitudinal analysis also demonstrated that SARS-CoV-2-specific T cell responses waned significantly in 75% of CIs during the follow-up. Collectively, we provide a comprehensive characterization of the long-term memory T cell response in CIs, suggesting that robust SARS-CoV-2-specific T cell immunity post primary infection may be less durable than previously expected.
Collapse
Affiliation(s)
- Ziwei Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tiandan Xiang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Boyun Liang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Deng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuecheng Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuemei Feng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mirko Trilling
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kathrin Sutter
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ulf Dittmer
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
Jelen Ž, Kovač J, Rudolf R. Study of Gold Nanoparticles Conjugated with SARS-CoV-2 S1 Spike Protein Fragments. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2160. [PMID: 37570478 PMCID: PMC10421057 DOI: 10.3390/nano13152160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023]
Abstract
This study reports on the successful conjugation of SARS-CoV-2 S1 spike protein fragments with gold nanoparticles (AuNPs) that were synthesised with Ultrasonic Spray Pyrolysis (USP). This method enables the continuous synthesis of AuNPs with a high degree of purity, round shapes, and the formation of a surface that allows various modifications. The conjugation mechanism of USP synthesized AuNPs with SARS-CoV-2 S1 spike protein fragments was investigated. A gel electrophoresis experiment confirmed the successful conjugation of AuNPs with SARS-CoV-2 S1 fragments indirectly. X-ray Photoelectron Spectroscopy (XPS) analysis confirmed the presence of characteristic O1s and N1s peaks, which indicated that specific binding between AuNPs and SARS-CoV-2 S1 spike protein fragments takes place via a peptide bond formed with the citrate stabiliser. This bond is coordinated to the AuNP's surface and the N-terminals of the protein, with the conjugate displaying the expected response within a prototype LFIA test. This study will help in better understanding the behaviour of AuNPs synthesised with USP and their potential use as sensors in colorimetric or electrochemical sensors and LFIA tests.
Collapse
Affiliation(s)
- Žiga Jelen
- Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia;
| | - Janez Kovač
- Department of Surface Engineering, Institut Jožef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia;
| | - Rebeka Rudolf
- Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia;
| |
Collapse
|
28
|
Hamdy ME, El Deeb AH, Hagag NM, Shahein MA, Alaidi O, Hussein HA. Interspecies transmission of SARS CoV-2 with special emphasis on viral mutations and ACE-2 receptor homology roles. Int J Vet Sci Med 2023; 11:55-86. [PMID: 37441062 PMCID: PMC10334861 DOI: 10.1080/23144599.2023.2222981] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
COVID-19 outbreak was first reported in 2019, Wuhan, China. The spillover of the disease caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), to a wide range of pet, zoo, wild, and farm animals has emphasized potential zoonotic and reverse zoonotic viral transmission. Furthermore, it has evoked inquiries about susceptibility of different animal species to SARS-CoV-2 infection and role of these animals as viral reservoirs. Therefore, studying susceptible and non-susceptible hosts for SARS-CoV-2 infection could give a better understanding for the virus and will help in preventing further outbreaks. Here, we review structural aspects of SARS-CoV-2 spike protein, the effect of the different mutations observed in the spike protein, and the impact of ACE2 receptor variations in different animal hosts on inter-species transmission. Moreover, the SARS-CoV-2 spillover chain was reviewed. Combination of SARS-CoV-2 high mutation rate and homology of cellular ACE2 receptors enable the virus to transcend species barriers and facilitate its transmission between humans and animals.
Collapse
Affiliation(s)
- Mervat E. Hamdy
- Genome Research Unit, Animal Health Research Institute, Agriculture Research Centre, Giza, Egypt
| | - Ayman H. El Deeb
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Department of Virology, Faculty of Veterinary Medicine, King Salman International University, South Sinai, Egypt
| | - Naglaa M. Hagag
- Genome Research Unit, Animal Health Research Institute, Agriculture Research Centre, Giza, Egypt
| | - Momtaz A. Shahein
- Department of Virology, Animal Health Research Institute, Agriculture Research Centre, Giza, Egypt
| | - Osama Alaidi
- Biocomplexity for Research and Consulting Co., Cairo, Egypt
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hussein A. Hussein
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
29
|
Mei X, Li J, Wang Z, Zhu D, Huang K, Hu S, Popowski KD, Cheng K. An inhaled bioadhesive hydrogel to shield non-human primates from SARS-CoV-2 infection. NATURE MATERIALS 2023; 22:903-912. [PMID: 36759564 PMCID: PMC10615614 DOI: 10.1038/s41563-023-01475-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
The surge of fast-spreading SARS-CoV-2 mutated variants highlights the need for fast, broad-spectrum strategies to counteract viral infections. In this work, we report a physical barrier against SARS-CoV-2 infection based on an inhalable bioadhesive hydrogel, named spherical hydrogel inhalation for enhanced lung defence (SHIELD). Conveniently delivered via a dry powder inhaler, SHIELD particles form a dense hydrogel network that coats the airway, enhancing the diffusional barrier properties and restricting virus penetration. SHIELD's protective effect is first demonstrated in mice against two SARS-CoV-2 pseudo-viruses with different mutated spike proteins. Strikingly, in African green monkeys, a single SHIELD inhalation provides protection for up to 8 hours, efficiently reducing infection by the SARS-CoV-2 WA1 and B.1.617.2 (Delta) variants. Notably, SHIELD is made with food-grade materials and does not affect normal respiratory functions. This approach could offer additional protection to the population against SARS-CoV-2 and other respiratory pathogens.
Collapse
Affiliation(s)
- Xuan Mei
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Junlang Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Zhenzhen Wang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Kristen D Popowski
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA.
| |
Collapse
|
30
|
Patel RS, Agrawal B. Mucosal immunization with lipopeptides derived from conserved regions of SARS-CoV-2 antigens induce robust cellular and cross-variant humoral immune responses in mice. Front Immunol 2023; 14:1178523. [PMID: 37334376 PMCID: PMC10272440 DOI: 10.3389/fimmu.2023.1178523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/17/2023] [Indexed: 06/20/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, has infected >600 million people in the ongoing global pandemic. Several variants of the SARS-CoV-2 have emerged in the last >2 years, challenging the continued efficacy of current COVID vaccines. Therefore, there is a crucial need to investigate a highly cross-protective vaccine effective against variants of SARS-CoV-2. In this study, we examined seven lipopeptides derived from highly conserved, immunodominant epitopes from the S, N, and M proteins of SARS-CoV-2, that are predicted to contain epitopes for clinically protective B cells, helper T cells (TH) and cytotoxic T cells (CTL). Intranasal immunization of mice with most of the lipopeptides led to significantly higher splenocyte proliferation and cytokine production, mucosal and systemic antibody responses, and induction of effector B and T lymphocytes in both lungs and spleen, compared to immunizations with the corresponding peptides without lipid. Immunizations with Spike-derived lipopeptides led to cross-reactive IgG, IgM and IgA responses against Alpha, Beta, Delta, and Omicron Spike proteins as well as neutralizing antibodies. These studies support their potential for development as components of a cross-protective SARS-CoV-2 vaccine.
Collapse
|
31
|
García-Bernalt Diego J, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian Hamsters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541294. [PMID: 37425792 PMCID: PMC10327228 DOI: 10.1101/2023.05.22.541294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Hybrid immunity to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Spain
- 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
| | - 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
| | - Sonia Jangra
- 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
| | - Kim Handrejk
- 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
| | - Manon Laporte
- 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
| | - 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
| | - Sara S El Zahed
- 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
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Max W Chang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - 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
| | - Sadaf Aslam
- 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
| | - Ignacio Mena
- 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
| | - Brett T Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, USA
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, CA, 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 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
| |
Collapse
|
32
|
Tworek A, Jaroń K, Cicha M, Rydzewski A, Wierzba W, Zaczyński A, Król Z, Rydzewska G. The persistence of SARS-CoV-2 neutralizing antibodies after COVID-19: A one-year observation. Is a SARS-CoV-2 vaccination booster dose necessary? Cent Eur J Immunol 2023; 48:92-96. [PMID: 37692027 PMCID: PMC10485689 DOI: 10.5114/ceji.2023.126206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/27/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction The aim of this study was to investigate the persistence of SARS-CoV-2 neutralizing antibodies (NAbs) one year after contracting COVID-19. Material and methods The study included 38 patients - 34 men and 4 women - suffering from COVID-19 between March 15 and May 26, 2020. The median age in the group was 31 years, ranging from 22 to 67 years. The levels of neutralizing antibodies were measured at three time-points - baseline, 6 months, and 12 months. The primary endpoint was a post-infection positive result for NAbs (> 15 AU/ml; Liaison SARS-CoV-2 S1/S2 IgG quantitative test) 12 months after infection. Results The median level of NAbs after 12 months was 26.5 AU/ml. At the end of observation (12 months), 21 of the 38 patients had a NAb level of >15 AU/ml (positive). The median antibody half-life was 5.8 months. Conclusions A high percentage of the patients maintained positive levels of antibodies 6 and 12 months after COVID-19 infection. The dynamics of the antibody level decline suggests the need for booster vaccination at least once a year.
Collapse
Affiliation(s)
- Adam Tworek
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Krzysztof Jaroń
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Małgorzata Cicha
- Diagnostic Laboratory of Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Andrzej Rydzewski
- Department of Internal Medicine, Nephrology and Transplantation Medicine, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
- Department of Internal Medicine, Nephrology and Transplantation Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Waldemar Wierzba
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
- University of Humanities and Economics in Łódz, Satellite Campus in Warsaw, Warsaw, Poland
| | - Artur Zaczyński
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Zbigniew Król
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Grażyna Rydzewska
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
33
|
Triambak S, Mahapatra D, Barik N, Chutjian A. Plausible explanation for the third COVID-19 wave in India and its implications. Infect Dis Model 2023; 8:183-191. [PMID: 36643865 PMCID: PMC9824946 DOI: 10.1016/j.idm.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023] Open
Abstract
Recently some of us used a random-walk Monte Carlo simulation approach to study the spread of COVID-19. The calculations were reasonably successful in describing secondary and tertiary waves of infection, in countries such as the USA, India, South Africa and Serbia. However, they failed to predict the observed third wave for India. In this work we present a more complete set of simulations for India, that take into consideration two aspects that were not incorporated previously. These include the stochastic movement of an erstwhile protected fraction of the population, and the reinfection of some recovered individuals because of their exposure to a new variant of the SARS-CoV-2 virus. The extended simulations now show the third COVID-19 wave for India that was missing in the earlier calculations. They also suggest an additional fourth wave, which was indeed observed during approximately the same time period as the model prediction.
Collapse
Affiliation(s)
- S. Triambak
- Department of Physics and Astronomy, University of the Western Cape, P/B X17, Bellville, 7535, South Africa,Corresponding author
| | - D.P. Mahapatra
- Department of Physics, Utkal University, Vani Vihar, Bhubaneshwar, 751004, India
| | - N. Barik
- Department of Physics, Utkal University, Vani Vihar, Bhubaneshwar, 751004, India
| | - A. Chutjian
- Armenian Engineers and Scientists of America, 326 Mira Loma Ave., Glendale, CA, 91204, USA
| |
Collapse
|
34
|
Graninger M, Jani CM, Reuberger E, Prüger K, Gaspar P, Springer DN, Borsodi C, Weidner L, Rabady S, Puchhammer-Stöckl E, Jungbauer C, Höltl E, Aberle JH, Stiasny K, Weseslindtner L. Comprehensive Comparison of Seven SARS-CoV-2-Specific Surrogate Virus Neutralization and Anti-Spike IgG Antibody Assays Using a Live-Virus Neutralization Assay as a Reference. Microbiol Spectr 2023; 11:e0231422. [PMID: 36622205 PMCID: PMC9927416 DOI: 10.1128/spectrum.02314-22] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Neutralizing antibodies (nAbs) are considered a valuable marker for measuring humoral immunity against SARS-CoV-2. However, live-virus neutralization tests (NTs) require high-biosafety-level laboratories and are time-consuming. Therefore, surrogate virus neutralization tests (sVNTs) have been widely applied, but unlike most anti-spike (S) antibody assays, NTs and sVNTs are not harmonized, requiring further evaluation and comparative analyses. This study compared seven commercial sVNTs and anti-S-antibody assays with a live-virus NT as a reference, using a panel of 720 single and longitudinal serum samples from 666 convalescent patients after SARS-CoV-2 infection. The sensitivity of these assays for detecting antibodies ranged from 48 to 94% after PCR-confirmed infection and from 56% to 100% relative to positivity in the in-house live-virus NT. Furthermore, we performed receiver operating characteristic (ROC) curve analyses to determine which immunoassays were most suitable for assessing nAb titers exceeding a specific cutoff (NT titer, ≥80) and found that the NeutraLISA and the cPass assays reached the highest area under the curve (AUC), exceeding 0.91. In addition, when the assays were compared for their correlation with nAb kinetics over time in a set of longitudinal samples, the extent of the measured decrease of nAbs after infection varied widely among the evaluated immunoassays. Finally, in vaccinated convalescent patients, high titers of nAbs exceeded the upper limit of the evaluated assays' quantification ranges. Based on data from this study, we conclude that commercial immunoassays are acceptable substitutes for live-virus NTs, particularly when additional adapted cutoffs are employed to detect nAbs beyond a specific threshold titer. IMPORTANCE While the measurement of neutralizing antibodies is considered a valuable tool in assessing protection against SARS-CoV-2, neutralization tests employ live-virus isolates and cell culture, requiring advanced laboratory biosafety levels. Including a large sample panel (over 700 samples), this study provides adapted cutoff values calculated for seven commercial immunoassays (including four surrogate neutralization assays and a protein-based microarray) that robustly correlate with specific titers of neutralizing antibodies.
Collapse
Affiliation(s)
| | | | | | - Katja Prüger
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Philipp Gaspar
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | | | - Lisa Weidner
- Austrian Red Cross, Blood Service for Vienna, Lower Austria, and Burgenland, Vienna, Austria
| | - Susanne Rabady
- Karl Landsteiner University of Health Sciences, Department of General Health Studies, Division General and Family Medicine, Krems, Austria
| | | | - Christof Jungbauer
- Austrian Red Cross, Blood Service for Vienna, Lower Austria, and Burgenland, Vienna, Austria
| | - Eva Höltl
- Center for Public Health, Medical University of Vienna, Vienna, Austria
| | | | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
35
|
Characterization of a Vesicular Stomatitis Virus-Vectored Recombinant Virus Bearing Spike Protein of SARS-CoV-2 Delta Variant. Microorganisms 2023; 11:microorganisms11020431. [PMID: 36838396 PMCID: PMC9960918 DOI: 10.3390/microorganisms11020431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
The frequent emergence of SARS-CoV-2 variants thwarts the prophylactic and therapeutic countermeasures confronting COVID-19. Among them, the Delta variant attracts widespread attention due to its high pathogenicity and fatality rate compared with other variants. However, with the emergence of new variants, studies on Delta variants have been gradually weakened and ignored. In this study, a replication-competent recombinant virus carrying the S protein of the SARS-CoV-2 Delta variant was established based on the vesicular stomatitis virus (VSV), which presented a safe alternative model for studying the Delta variant. The recombinant virus showed a replication advantage in Vero E6 cells, and the viral titers reach 107.3 TCID50/mL at 36 h post-inoculation. In the VSV-vectored recombinant platform, the spike proteins of the Delta variant mediated higher fusion activity and syncytium formation than the wild-type strain. Notably, the recombinant virus was avirulent in BALB/c mice, Syrian hamsters, 3-day ICR suckling mice, and IFNAR/GR-/- mice. It induced protective neutralizing antibodies in rodents, and protected the Syrian hamsters against the SARS-CoV-2 Delta variant infection. Meanwhile, the eGFP reporter of recombinant virus enabled the visual assay of neutralizing antibodies. Therefore, the recombinant virus could be a safe and convenient surrogate tool for authentic SARS-CoV-2. This efficient and reliable model has significant potential for research on viral-host interactions, epidemiological investigation of serum-neutralizing antibodies, and vaccine development.
Collapse
|
36
|
Dillard JA, Martinez SA, Dearing JJ, Montgomery SA, Baxter AK. Animal Models for the Study of SARS-CoV-2-Induced Respiratory Disease and Pathology. Comp Med 2023; 73:72-90. [PMID: 36229170 PMCID: PMC9948904 DOI: 10.30802/aalas-cm-22-000089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emergence of the betacoronavirus SARS-CoV-2 has resulted in a historic pandemic, with millions of deaths worldwide. An unprecedented effort has been made by the medical, scientific, and public health communities to rapidly develop and implement vaccines and therapeutics to prevent and reduce hospitalizations and deaths. Although SARS-CoV-2 infection can lead to disease in many organ systems, the respiratory system is its main target, with pneumonia and acute respiratory distress syndrome as the hallmark features of severe disease. The large number of patients who have contracted COVID-19 infections since 2019 has permitted a detailed characterization of the clinical and pathologic features of the disease in humans. However, continued progress in the development of effective preventatives and therapies requires a deeper understanding of the pathogenesis of infection. Studies using animal models are necessary to complement in vitro findings and human clinical data. Multiple animal species have been evaluated as potential models for studying the respiratory disease caused by SARSCoV-2 infection. Knowing the similarities and differences between animal and human responses to infection is critical for effective translation of animal data into human medicine. This review provides a detailed summary of the respiratory disease and associated pathology induced by SARS-CoV-2 infection in humans and compares them with the disease that develops in 3 commonly used models: NHP, hamsters, and mice. The effective use of animals to study SARS-CoV-2-induced respiratory disease will enhance our understanding of SARS-CoV-2 pathogenesis, allow the development of novel preventatives and therapeutics, and aid in the preparation for the next emerging virus with pandemic potential.
Collapse
Key Words
- ace2, angiotensin-converting enzyme 2
- agm, african green monkey
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- balf, bronchoalveolar lavage fluid
- cards, covid-19-associated acute respiratory distress syndrome
- dad, diffuse alveolar damage
- dpi, days postinfection
- ggo, ground glass opacities
- s, spike glycoprotein
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sabian A Martinez
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Justin J Dearing
- Biological and Biomedical Sciences Program, Office of Graduate Education, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andvictoria K Baxter
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;,
| |
Collapse
|
37
|
mRNA-From COVID-19 Treatment to Cancer Immunotherapy. Biomedicines 2023; 11:biomedicines11020308. [PMID: 36830845 PMCID: PMC9953480 DOI: 10.3390/biomedicines11020308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
This review provides an overview covering mRNA from its use in the COVID-19 pandemic to cancer immunotherapy, starting from the selection of appropriate antigens, tumor-associated and tumor-specific antigens, neoantigens, the basics of optimizing the mRNA molecule in terms of stability, efficacy, and tolerability, choosing the best formulation and the optimal route of administration, to summarizing current clinical trials of mRNA vaccines in tumor therapy.
Collapse
|
38
|
Abebe EC, Dejenie TA. Protective roles and protective mechanisms of neutralizing antibodies against SARS-CoV-2 infection and their potential clinical implications. Front Immunol 2023; 14:1055457. [PMID: 36742320 PMCID: PMC9892939 DOI: 10.3389/fimmu.2023.1055457] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Neutralizing antibodies (NAbs) are central players in the humoral immunity that defends the body from SARS-CoV-2 infection by blocking viral entry into host cells and neutralizing their biological effects. Even though NAbs primarily work by neutralizing viral antigens, on some occasions, they may also combat the SARS-CoV-2 virus escaping neutralization by employing several effector mechanisms in collaboration with immune cells like natural killer (NK) cells and phagocytes. Besides their prophylactic and therapeutic roles, antibodies can be used for COVID-19 diagnosis, severity evaluation, and prognosis assessment in clinical practice. Furthermore, the measurement of NAbs could have key implications in determining individual or herd immunity against SARS-CoV-2, vaccine effectiveness, and duration of the humoral protective response, as well as aiding in the selection of suitable individuals who can donate convalescent plasma to treat infected people. Despite all these clinical applications of NAbs, using them in clinical settings can present some challenges. This review discusses the protective functions, possible protective mechanisms against SARS-CoV-2, and potential clinical applications of NAbs in COVID-19. This article also highlights the possible challenges and solutions associated with COVID-19 antibody-based prophylaxis, therapy, and vaccination.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
39
|
Tyrkalska SD, Candel S, Pedoto A, García-Moreno D, Alcaraz-Pérez F, Sánchez-Ferrer Á, Cayuela ML, Mulero V. Zebrafish models of COVID-19. FEMS Microbiol Rev 2023; 47:fuac042. [PMID: 36323404 PMCID: PMC9841970 DOI: 10.1093/femsre/fuac042] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Although COVID-19 has only recently appeared, research studies have already developed and implemented many animal models for deciphering the secrets of the disease and provided insights into the biology of SARS-CoV-2. However, there are several major factors that complicate the study of this virus in model organisms, such as the poor infectivity of clinical isolates of SARS-CoV-2 in some model species, and the absence of persistent infection, immunopathology, severe acute respiratory distress syndrome, and, in general, all the systemic complications which characterize COVID-19 clinically. Another important limitation is that SARS-CoV-2 mainly causes severe COVID-19 in older people with comorbidities, which represents a serious problem when attempting to use young and immunologically naïve laboratory animals in COVID-19 testing. We review here the main animal models developed so far to study COVID-19 and the unique advantages of the zebrafish model that may help to contribute to understand this disease, in particular to the identification and repurposing of drugs to treat COVID-19, to reveal the mechanism of action and side-effects of Spike-based vaccines, and to decipher the high susceptibility of aged people to COVID-19.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Candel
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Annamaria Pedoto
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Diana García-Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisca Alcaraz-Pérez
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Telomerasa, Cáncer y Envejecimiento (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Álvaro Sánchez-Ferrer
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Departmento de Bioloquímica y Biología Molecular A, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | - María L Cayuela
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Telomerasa, Cáncer y Envejecimiento (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
40
|
Porter AF, Purcell DFJ, Howden BP, Duchene S. Evolutionary rate of SARS-CoV-2 increases during zoonotic infection of farmed mink. Virus Evol 2023; 9:vead002. [PMID: 36751428 PMCID: PMC9896948 DOI: 10.1093/ve/vead002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/11/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
To investigate genetic signatures of adaptation to the mink host, we characterised the evolutionary rate heterogeneity in mink-associated severe acute respiratory syndrome coronaviruses (SARS-CoV-2). In 2020, the first detected anthropozoonotic spillover event of SARS-CoV-2 occurred in mink farms throughout Europe and North America. Both spill-back of mink-associated lineages into the human population and the spread into the surrounding wildlife were reported, highlighting the potential formation of a zoonotic reservoir. Our findings suggest that the evolutionary rate of SARS-CoV-2 underwent an episodic increase upon introduction into the mink host before returning to the normal range observed in humans. Furthermore, SARS-CoV-2 lineages could have circulated in the mink population for a month before detection, and during this period, evolutionary rate estimates were between 3 × 10-3 and 1.05 × 10-2 (95 per cent HPD, with a mean rate of 6.59 × 10-3) a four- to thirteen-fold increase compared to that in humans. As there is evidence for unique mutational patterns within mink-associated lineages, we explored the emergence of four mink-specific Spike protein amino acid substitutions Y453F, S1147L, F486L, and Q314K. We found that mutation Y453F emerged early in multiple mink outbreaks and that mutations F486L and Q314K may co-occur. We suggest that SARS-CoV-2 undergoes a brief, but considerable, increase in evolutionary rate in response to greater selective pressures during species jumps, which may lead to the occurrence of mink-specific mutations. These findings emphasise the necessity of ongoing surveillance of zoonotic SARS-CoV-2 infections in the future.
Collapse
Affiliation(s)
- Ashleigh F Porter
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Damian F J Purcell
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Benjamin P Howden
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
- Microbiological Diagnostic Unit Public Health Laboratory, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sebastian Duchene
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
41
|
Refocus on Immunogenic Characteristics of Convalescent COVID-19 Challenged by Prototype SARS-CoV-2. Vaccines (Basel) 2023; 11:vaccines11010123. [PMID: 36679968 PMCID: PMC9866260 DOI: 10.3390/vaccines11010123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Background: Mass basic and booster immunization programs effectively contained the spread of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus, also known as COVID-19. However, the emerging Variants of Concern (VOCs) of COVID-19 evade the immune protection of the vaccine and increase the risk of reinfection. Methods: Serum antibodies of 384 COVID-19 cases recovered from SARS-CoV-2 infection were examined. Correlations between clinical symptoms and antibodies against VOCs were analyzed. Result: All 384 cases (aged 43, range 1−90) were from 15 cities of Guangdong, China. The specific IgA, IgG, and IgM antibodies could be detected within 4−6 weeks after infection. A broad cross-reaction between SARS-CoV-2 and Severe Acute Respiratory Syndrome Coronavirus, but not with Middle East Respiratory Syndrome Coronavirus was found. The titers of neutralization antibodies (NAbs) were significantly correlated with IgG (r = 0.667, p < 0.001), but showed poor neutralizing effects against VOCs. Age, fever, and hormone therapy were independent risk factors for NAbs titers reduction against VOCs. Conclusion: Humoral immunity antibodies from the original strain of COVID-19 showed weak neutralization effects against VOCs, and decreased neutralizing ability was associated with initial age, fever, and hormone therapy, which hindered the effects of the COVID-19 vaccine developed from the SARS-CoV-2 prototype virus.
Collapse
|
42
|
LÉCU ALEXIS. SARS-Cov-2 Coronavirus Infection in Wild Animals. FOWLER' S ZOO AND WILD ANIMAL MEDICINE CURRENT THERAPY, VOLUME 10 2023:113-120. [DOI: 10.1016/b978-0-323-82852-9.00018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Tailor N, Warner BM, Griffin BD, Tierney K, Moffat E, Frost K, Vendramelli R, Leung A, Willman M, Thomas SP, Pei Y, Booth SA, Embury-Hyatt C, Wootton SK, Kobasa D. Generation and Characterization of a SARS-CoV-2-Susceptible Mouse Model Using Adeno-Associated Virus (AAV6.2FF)-Mediated Respiratory Delivery of the Human ACE2 Gene. Viruses 2022; 15:85. [PMID: 36680125 PMCID: PMC9863330 DOI: 10.3390/v15010085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the aetiological agent of coronavirus disease 2019 (COVID-19) that has caused a pandemic with millions of human infections. There continues to be a pressing need to develop potential therapies and vaccines to inhibit SARS-CoV-2 infection to mitigate the ongoing pandemic. Epidemiological data from the current pandemic indicates that there may be sex-dependent differences in disease outcomes. To investigate these differences, we proposed to use common small animal species that are frequently used to model disease with viruses. However, common laboratory strains of mice are not readily infected by SARS-CoV-2 because of differences in the angiotensin-converting enzyme 2 (ACE2), the cellular receptor for the virus. To overcome this limitation, we transduced common laboratory accessible strains of mice of different sexes and age groups with a novel a triple AAV6 mutant, termed AAV6.2FF, encoding either human ACE2 or luciferase via intranasal administration to promote expression in the lung and nasal turbinates. Infection of AAV-hACE2-transduced mice with SARS-CoV-2 resulted in high viral titers in the lungs and nasal turbinates, establishment of an IgM and IgG antibody response, and modulation of lung and nasal turbinate cytokine profiles. There were insignificant differences in infection characteristics between age groups and sex-related differences; however, there were significant strain-related differences between BALB/c vs. C57BL/6 mice. We show that AAV-hACE2-transduced mice are a useful for determining immune responses and for potential evaluation of SARS-CoV-2 vaccines and antiviral therapies, and this study serves as a model for the utility of this approach to rapidly develop small-animal models for emerging viruses.
Collapse
Affiliation(s)
- Nikesh Tailor
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Bryce M. Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Bryan D. Griffin
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Kevin Tierney
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Estella Moffat
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, 1015 Arlington Street, Winnipeg, MB R3E 3M4, Canada
| | - Kathy Frost
- Molecular Pathobiology, National Microbiology Laboratory NML, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Robert Vendramelli
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Anders Leung
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
| | - Marnie Willman
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, College of Medicine, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| | - Sylvia P. Thomas
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Stephanie A. Booth
- Molecular Pathobiology, National Microbiology Laboratory NML, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, College of Medicine, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, 1015 Arlington Street, Winnipeg, MB R3E 3M4, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, College of Medicine, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
44
|
Blockchain in Healthcare: A Decentralized Platform for Digital Health Passport of COVID-19 Based on Vaccination and Immunity Certificates. Healthcare (Basel) 2022; 10:healthcare10122453. [PMID: 36553977 PMCID: PMC9778149 DOI: 10.3390/healthcare10122453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19 has become a very transmissible disease that has had a worldwide impact, resulting in a huge number of infections and fatalities. Testing is critical to the pandemic's successful response because it helps detect illnesses and so attenuate (isolate/cure) them and now vaccination is a life-safer innovation against the pandemic which helps to make the immunity system stronger and fight against this infection. Patient-sensitive information, on the other hand, is now held in a centralized or third-party storage paradigm, according to COVID-19. One of the most difficult aspects of using a centralized storage strategy is maintaining patient privacy and system transparency. The application of blockchain technology to support health initiatives that can minimize the spread of COVID-19 infections in the context of accessibility of the system and for verification of digital passports. Only by combining blockchain technology with advanced cryptographic algorithms can a secure and privacy-preserving solution to COVID-19 be provided. In this article, we investigate the issue and propose a blockchain-based solution incorporating conscience identity, encryption, and decentralized storage via interplanetary file systems (IPFS). For COVID-19 test takers and vaccination takers, our solution includes digital health passports (DHP) as a certification of test or vaccination. We explain smart contracts constructed and tested with Ethereum to preserve a DHP for test and vaccine takers, allowing for a prompt and trustworthy response from the necessary medical authorities. We use an immutable trustworthy blockchain to minimize medical facility response times, relieve the transmission of incorrect information, and stop the illness from spreading via DHP. We give a detailed explanation of the proposed solution's system model, development, and assessment in terms of cost and security. Finally, we put the suggested framework to the test by deploying a smart contract prototype on the Ethereum TESTNET network in a Windows environment. The study's findings revealed that the suggested method is effective and feasible.
Collapse
|
45
|
Differential transcriptomic landscapes of multiple organs from SARS-CoV-2 early infected rhesus macaques. Protein Cell 2022; 13:920-939. [PMID: 35377064 PMCID: PMC8978510 DOI: 10.1007/s13238-022-00915-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 infection causes complicated clinical manifestations with variable multi-organ injuries, however, the underlying mechanism, in particular immune responses in different organs, remains elusive. In this study, comprehensive transcriptomic alterations of 14 tissues from rhesus macaque infected with SARS-CoV-2 were analyzed. Compared to normal controls, SARS-CoV-2 infection resulted in dysregulation of genes involving diverse functions in various examined tissues/organs, with drastic transcriptomic changes in cerebral cortex and right ventricle. Intriguingly, cerebral cortex exhibited a hyperinflammatory state evidenced by significant upregulation of inflammation response-related genes. Meanwhile, expressions of coagulation, angiogenesis and fibrosis factors were also up-regulated in cerebral cortex. Based on our findings, neuropilin 1 (NRP1), a receptor of SARS-CoV-2, was significantly elevated in cerebral cortex post infection, accompanied by active immune response releasing inflammatory factors and signal transmission among tissues, which enhanced infection of the central nervous system (CNS) in a positive feedback way, leading to viral encephalitis. Overall, our study depicts a multi-tissue/organ transcriptomic landscapes of rhesus macaque with early infection of SARS-CoV-2, and provides important insights into the mechanistic basis for COVID-19-associated clinical complications.
Collapse
|
46
|
Subgenomic RNAs and Their Encoded Proteins Contribute to the Rapid Duplication of SARS-CoV-2 and COVID-19 Progression. Biomolecules 2022; 12:biom12111680. [DOI: 10.3390/biom12111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is currently widespread throughout the world, accompanied by a rising number of people infected and breakthrough infection of variants, which make the virus highly transmissible and replicable. A comprehensive understanding of the molecular virological events and induced immunological features during SARS-CoV-2 replication can provide reliable targets for vaccine and drug development. Among the potential targets, subgenomic RNAs and their encoded proteins involved in the life cycle of SARS-CoV-2 are extremely important in viral duplication and pathogenesis. Subgenomic RNAs employ a range of coping strategies to evade immune surveillance from replication to translation, which allows RNAs to synthesize quickly, encode structural proteins efficiently and complete the entire process of virus replication and assembly successfully. This review focuses on the characteristics and functions of SARS-CoV-2 subgenomic RNAs and their encoded proteins and explores in depth the role of subgenomic RNAs in the replication and infection of host cells to provide important clues to the mechanism of COVID-19 pathogenesis.
Collapse
|
47
|
Liu S, Stauft CB, Selvaraj P, Chandrasekaran P, D’Agnillo F, Chou CK, Wu WW, Lien CZ, Meseda CA, Pedro CL, Starost MF, Weir JP, Wang TT. Intranasal delivery of a rationally attenuated SARS-CoV-2 is immunogenic and protective in Syrian hamsters. Nat Commun 2022; 13:6792. [PMID: 36357440 PMCID: PMC9648440 DOI: 10.1038/s41467-022-34571-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Few live attenuated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are in pre-clinical or clinical development. We seek to attenuate SARS-CoV-2 (isolate WA1/2020) by removing the polybasic insert within the spike protein and the open reading frames (ORFs) 6-8, and by introducing mutations that abolish non-structural protein 1 (Nsp1)-mediated toxicity. The derived virus (WA1-ΔPRRA-ΔORF6-8-Nsp1K164A/H165A) replicates to 100- to 1000-fold-lower titers than the ancestral virus and induces little lung pathology in both K18-human ACE2 (hACE2) transgenic mice and Syrian hamsters. Immunofluorescence and transcriptomic analyses of infected hamsters confirm that three-pronged genetic modifications attenuate the proinflammatory pathways more than the removal of the polybasic cleavage site alone. Finally, intranasal administration of just 100 PFU of the WA1-ΔPRRA-ΔORF6-8-Nsp1K164A/H165A elicits robust antibody responses in Syrian hamsters and protects against SARS-CoV-2-induced weight loss and pneumonia. As a proof-of-concept study, we demonstrate that live but sufficiently attenuated SARS-CoV-2 vaccines may be attainable by rational design.
Collapse
Affiliation(s)
- Shufeng Liu
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Charles B. Stauft
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Prabhuanand Selvaraj
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Prabha Chandrasekaran
- grid.94365.3d0000 0001 2297 5165Laboratory of Clinical Investigation, National Institutes of Aging, National Institutes of Health, Baltimore, USA
| | - Felice D’Agnillo
- grid.417587.80000 0001 2243 3366Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Chao-Kai Chou
- grid.417587.80000 0001 2243 3366Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Wells W. Wu
- grid.417587.80000 0001 2243 3366Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Christopher Z. Lien
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Clement A. Meseda
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Cyntia L. Pedro
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Matthew F. Starost
- grid.94365.3d0000 0001 2297 5165Division of Veterinary Resources, Diagnostic and Research Services Branch, National Institutes of Health, Rockville Pike, USA
| | - Jerry P. Weir
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| | - Tony T. Wang
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD USA
| |
Collapse
|
48
|
A retrospective cross-sectional observational study of SARS-CoV-2 reinfection in La Ribera Health Department, Valencia, Spain. J Med Microbiol 2022; 71. [DOI: 10.1099/jmm.0.001599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. The possibility of reinfection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a widely proven fact and may have clinical implications.
Hypothesis /Gap Statement. It is not known whether there have been cases of reinfection by SARS-CoV-2 in La Ribera Health Department.
Aim. To determine whether there have been cases of reinfection by SARS-CoV-2 in La Ribera Health Department and to identify their characteristics.
Methodology. Retrospective cross-sectional observational study of cases of reinfection by SARS-CoV-2 in the population of La Ribera Department between March 2020 and February 2021. The positive baseline cohort includes all cases positive by RT-PCR for SARS-CoV-2, with reinfection cases being those that, after resolution of the first episode according to the World Health Organization (WHO) criteria, presented a new positive RT-PCR result.
Results. Out of a total of 15 687 cases with positive RT-PCR, 40 were considered to be reinfections, which meant a cumulative incidence of 0.255 % and an incidence density of 5.05 cases per 100 000 person-days. Most of the cases occurred during the highest incidence peaks of the pandemic in the department. Seventy-five per cent of the patients in these cases were older than 40 years, 42.5 % were healthcare professionals or nursing home residents and 12.5 % had an immunosuppressive comorbidity. There were no severe, critical or death cases. In the reinfection episodes, with respect to the first episode, there was a tendency to be milder, they required fewer days of hospitalization, their RT-PCR became negative earlier, they developed a greater humoral response and the sick leave period was shorter. The median period between the RT-PCR in the first episode and the RT-PCR in the second episode was 127.5 days (range: 48–301; IQR: 89.5–256.25)
Conclusions. SARS-CoV-2 reinfection cases are rare, tend to be mild and can occur within a median period of 127.5 days.
Collapse
|
49
|
Qi F, Qin C. Characteristics of animal models for COVID-19. Animal Model Exp Med 2022; 5:401-409. [PMID: 36301011 PMCID: PMC9610135 DOI: 10.1002/ame2.12278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), the most consequential pandemic of this century, threatening human health and public safety. SARS-CoV-2 has been continuously evolving through mutation of its genome and variants of concern have emerged. The World Health Organization R&D Blueprint plan convened a range of expert groups to develop animal models for COVID-19, a core requirement for the prevention and control of SARS-CoV-2 pandemic. The animal model construction techniques developed during the SARS-CoV and MERS-CoV pandemics were rapidly deployed and applied in the establishment of COVID-19 animal models. To date, a large number of animal models for COVID-19, including mice, hamsters, minks and nonhuman primates, have been established. Infectious diseases produce unique manifestations according to the characteristics of the pathogen and modes of infection. Here we classified animal model resources around the infection route of SARS-CoV-2, and summarized the characteristics of the animal models constructed via transnasal, localized, and simulated transmission routes of infection.
Collapse
Affiliation(s)
- Feifei Qi
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina,National Center of Technology Innovation for Animal ModelBeijingChina
| | - Chuan Qin
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina,National Center of Technology Innovation for Animal ModelBeijingChina
| |
Collapse
|
50
|
B.1.351 SARS-CoV-2 Variant Exhibits Higher Virulence but Less Viral Shedding than That of the Ancestral Strain in Young Nonhuman Primates. Microbiol Spectr 2022; 10:e0226322. [PMID: 36069561 PMCID: PMC9603226 DOI: 10.1128/spectrum.02263-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We investigated the distribution, virulence, and pathogenic characteristics of mutated SARS-CoV-2 to clarify the association between virulence and the viral spreading ability of current and future circulating strains. Chinese rhesus macaques were infected with ancestral SARS-CoV-2 strain GD108 and Beta variant B.1.351 (B.1.351) and assessed for clinical signs, viral distribution, pathological changes, and pulmonary inflammation. We found that GD108 replicated more efficiently in the upper respiratory tract, whereas B.1.351 replicated more efficiently in the lower respiratory tract and lung tissue, implying a reduced viral shedding and spreading ability of B.1.351 compared with that of GD108. Importantly, B.1.351 caused more severe lung injury and dramatically elevated the level of inflammatory cytokines compared with those observed after infection with GD108. Moreover, both B.1.351 and GD108 induced spike-specific T-cell responses at an early stage of infection, with higher levels of interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in the B.1.351 group and higher levels of interleukin 17 (IL-17) in the GD108 group, indicating a divergent pattern in the T-cell-mediated inflammatory "cytokine storm." This study provides a basis for exploring the pathogenesis of SARS-CoV-2 variants of concern (VOCs) and establishes an applicable animal model for evaluating the efficacy and safety of vaccines and drugs. IMPORTANCE One of the priorities of the current SARS-CoV-2 vaccine and drug research strategy is to determine the changes in transmission ability, virulence, and pathogenic characteristics of SARS-CoV-2 variants. In addition, nonhuman primates (NHPs) are suitable animal models for the study of the pathogenic characteristics of SARS-CoV-2 and could contribute to the understanding of pathogenicity and transmission mechanisms. As SARS-CoV-2 variants continually emerge and the viral biological characteristics change frequently, the establishment of NHP infection models for different VOCs is urgently needed. In the study, the virulence and tissue distribution of B.1.351 and GD108 were comprehensively studied in NHPs. We concluded that the B.1.351 strain was more virulent but exhibited less viral shedding than the latter. This study provides a basis for determining the pathogenic characteristics of SARS-CoV-2 and establishes an applicable animal model for evaluating the efficacy and safety of vaccines and drugs.
Collapse
|