1
|
Duan T, Xing C, Chu J, Deng X, Du Y, Liu X, Hu Y, Qian C, Yin B, Wang HY, Wang RF. ACE2-dependent and -independent SARS-CoV-2 entries dictate viral replication and inflammatory response during infection. Nat Cell Biol 2024; 26:628-644. [PMID: 38514841 DOI: 10.1038/s41556-024-01388-w] [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: 04/04/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Excessive inflammation is the primary cause of mortality in patients with severe COVID-19, yet the underlying mechanisms remain poorly understood. Our study reveals that ACE2-dependent and -independent entries of SARS-CoV-2 in epithelial cells versus myeloid cells dictate viral replication and inflammatory responses. Mechanistically, SARS-CoV-2 NSP14 potently enhances NF-κB signalling by promoting IKK phosphorylation, while SARS-CoV-2 ORF6 exerts an opposing effect. In epithelial cells, ACE2-dependent SARS-CoV-2 entry enables viral replication, with translated ORF6 suppressing NF-κB signalling. In contrast, in myeloid cells, ACE2-independent entry blocks the translation of ORF6 and other viral structural proteins due to inefficient subgenomic RNA transcription, but NSP14 could be directly translated from genomic RNA, resulting in an abortive replication but hyperactivation of the NF-κB signalling pathway for proinflammatory cytokine production. Importantly, we identified TLR1 as a critical factor responsible for viral entry and subsequent inflammatory response through interaction with E and M proteins, which could be blocked by the small-molecule inhibitor Cu-CPT22. Collectively, our findings provide molecular insights into the mechanisms by which strong viral replication but scarce inflammatory response during the early (ACE2-dependent) infection stage, followed by low viral replication and potent inflammatory response in the late (ACE2-independent) infection stage, may contribute to COVID-19 progression.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Junjun Chu
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiangxue Deng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xin Liu
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yuzhou Hu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chen Qian
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bingnan Yin
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Agrawal S, Tran MT, Jennings TSK, Soliman MMH, Heo S, Sasson B, Rahmatpanah F, Agrawal A. Changes in the innate immune response to SARS-CoV-2 with advancing age in humans. Immun Ageing 2024; 21:21. [PMID: 38515147 PMCID: PMC10956333 DOI: 10.1186/s12979-024-00426-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Advancing age is a major risk factor for respiratory viral infections. The infections are often prolonged and difficult to resolve resulting hospitalizations and mortality. The recent COVID-19 pandemic has highlighted this as elderly subjects have emerged as vulnerable populations that display increased susceptibility and severity to SARS-CoV-2. There is an urgent need to identify the probable mechanisms underlying this to protect against future outbreaks of such nature. Innate immunity is the first line of defense against viruses and its decline impacts downstream immune responses. This is because dendritic cells (DCs) and macrophages are key cellular elements of the innate immune system that can sense and respond to viruses by producing inflammatory mediators and priming CD4 and CD8 T-cell responses. RESULTS We investigated the changes in innate immune responses to SARS-CoV-2 as a function of age. Our results using human PBMCs from aged, middle-aged, and young subjects indicate that the activation of DCs and monocytes in response to SARS-CoV-2 is compromised with age. The impairment is most apparent in pDCs where both aged and middle-aged display reduced responses. The secretion of IL-29 that confers protection against respiratory viruses is also decreased in both aged and middle-aged subjects. In contrast, inflammatory mediators associated with severe COVID-19 including CXCL-8, TREM-1 are increased with age. This is also apparent in the gene expression data where pathways related host defense display an age dependent decrease with a concomitant increase in inflammatory pathways. Not only are the inflammatory pathways and mediators increased after stimulation with SARS-CoV-2 but also at homeostasis. In keeping with reduced DC activation, the induction of cytotoxic CD8 T cells is also impaired in aged subjects. However, the CD8 T cells from aged subjects display increased baseline activation in accordance with the enhanced baseline inflammation. CONCLUSIONS Our results demonstrate a decline in protective anti-viral immune responses and increase in damaging inflammatory responses with age indicating that dysregulated innate immune responses play a significant role in the increased susceptibility of aged subjects to COVID-19. Furthermore, the dysregulation in immune responses develops early on as middle-aged demonstrate several of these changes.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Michelle Thu Tran
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | | | - Marlaine Maged Hosny Soliman
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Sally Heo
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Bobby Sasson
- Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Farah Rahmatpanah
- Department of Pathology, University of California Irvine, Irvine, CA, 92697, USA
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
3
|
Chakraborty C, Bhattacharya M, Lee SS. Regulatory role of miRNAs in the human immune and inflammatory response during the infection of SARS-CoV-2 and other respiratory viruses: A comprehensive review. Rev Med Virol 2024; 34:e2526. [PMID: 38446531 DOI: 10.1002/rmv.2526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/11/2024] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
miRNAs are single-stranded ncRNAs that act as regulators of different human body processes. Several miRNAs have been noted to control the human immune and inflammatory response during severe acute respiratory infection syndrome (SARS-CoV-2) infection. Similarly, many miRNAs were upregulated and downregulated during different respiratory virus infections. Here, an attempt has been made to capture the regulatory role of miRNAs in the human immune and inflammatory response during the infection of SARS-CoV-2 and other respiratory viruses. Firstly, the role of miRNAs has been depicted in the human immune and inflammatory response during the infection of SARS-CoV-2. In this direction, several significant points have been discussed about SARS-CoV-2 infection, such as the role of miRNAs in human innate immune response; miRNAs and its regulation of granulocytes; the role of miRNAs in macrophage activation and polarisation; miRNAs and neutrophil extracellular trap formation; miRNA-related inflammatory response; and miRNAs association in adaptive immunity. Secondly, the miRNAs landscape has been depicted during human respiratory virus infections such as human coronavirus, respiratory syncytial virus, influenza virus, rhinovirus, and human metapneumovirus. The article will provide more understanding of the miRNA-controlled mechanism of the immune and inflammatory response during COVID-19, which will help more therapeutics discoveries to fight against the future pandemic.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| | | | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Gangwon-do, Republic of Korea
| |
Collapse
|
4
|
Mazzarella L, Santoro F, Ravasio R, Fumagalli V, Massa PE, Rodighiero S, Gavilán E, Romanenghi M, Duso BA, Bonetti E, Manganaro L, Pallavi R, Trastulli D, Pallavicini I, Gentile C, Monzani S, Leonardi T, Pasqualato S, Buttinelli G, Di Martino A, Fedele G, Schiavoni I, Stefanelli P, Meroni G, de Francesco R, Steinkuhler C, Fossati G, Iannacone M, Minucci S, Pelicci PG. Inhibition of the lysine demethylase LSD1 modulates the balance between inflammatory and antiviral responses against coronaviruses. Sci Signal 2023; 16:eade0326. [PMID: 38113337 DOI: 10.1126/scisignal.ade0326] [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: 07/20/2022] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
Innate immune responses to coronavirus infections are highly cell specific. Tissue-resident macrophages, which are infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in patients but are inconsistently infected in vitro, exert critical but conflicting effects by secreting both antiviral type I interferons (IFNs) and tissue-damaging inflammatory cytokines. Steroids, the only class of host-targeting drugs approved for the treatment of coronavirus disease 2019 (COVID-19), indiscriminately suppress both responses, possibly impairing viral clearance. Here, we established in vitro cell culture systems that enabled us to separately investigate the cell-intrinsic and cell-extrinsic proinflammatory and antiviral activities of mouse macrophages infected with the prototypical murine coronavirus MHV-A59. We showed that the nuclear factor κB-dependent inflammatory response to viral infection was selectively inhibited by loss of the lysine demethylase LSD1, which was previously implicated in innate immune responses to cancer, with negligible effects on the antiviral IFN response. LSD1 ablation also enhanced an IFN-independent antiviral response, blocking viral egress through the lysosomal pathway. The macrophage-intrinsic antiviral and anti-inflammatory activity of Lsd1 inhibition was confirmed in vitro and in a humanized mouse model of SARS-CoV-2 infection. These results suggest that LSD1 controls innate immune responses against coronaviruses at multiple levels and provide a mechanistic rationale for potentially repurposing LSD1 inhibitors for COVID-19 treatment.
Collapse
Affiliation(s)
- Luca Mazzarella
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Fabio Santoro
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Ravasio
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Milan 20132, Italy
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Paul E Massa
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Gavilán
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Mauro Romanenghi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Bruno A Duso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Emanuele Bonetti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Lara Manganaro
- Virology, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi," 20122 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Deborah Trastulli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Isabella Pallavicini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Gentile
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Silvia Monzani
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Sebastiano Pasqualato
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Gabriele Buttinelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Angela Di Martino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giorgio Fedele
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ilaria Schiavoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Meroni
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Raffaele de Francesco
- Virology, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi," 20122 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Christian Steinkuhler
- Preclinical R&D Italfarmaco SpA, Via dei Lavoratori 54, 20092 Cinisello Balsamo (Milan), Italy
| | - Gianluca Fossati
- Preclinical R&D Italfarmaco SpA, Via dei Lavoratori 54, 20092 Cinisello Balsamo (Milan), Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Milan 20132, Italy
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Biosciences, University of Milan, Milan 20123, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan 20122, Italy
| |
Collapse
|
5
|
Singh R, Malik P, Kumar M, Kumar R, Alam MS, Mukherjee TK. Secondary fungal infections in SARS-CoV-2 patients: pathological whereabouts, cautionary measures, and steadfast treatments. Pharmacol Rep 2023:10.1007/s43440-023-00506-z. [PMID: 37354313 DOI: 10.1007/s43440-023-00506-z] [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: 02/09/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
The earliest documented COVID-19 case caused by the SARS-CoV-2 coronavirus occurred in Wuhan, China, in December 2019. Since then, several SARS-CoV-2 mutants have rapidly disseminated as exemplified by the community spread of the recent omicron variant. The disease already attained a pandemic status with ever-dwindling mortality even after two and half years of identification and considerable vaccination. Aspergillosis, candidiasis, cryptococcosis and mucormycosis are the prominent fungal infections experienced by the majority of SARS-CoV-2 high-risk patients. In its entirety, COVID-19's nexus with these fungal infections may worsen the intricacies in the already beleaguered high-risk patients, making this a topic of substantial clinical concern. Thus, thorough knowledge of the subject is necessary. This article focuses on the concomitant fungal infection(s) in COVID-19 patients, taking into account their underlying causes, the screening methods, manifested drug resistance, and long-term effects. The information and knowledge shared herein could be crucial for the management of critically ill, aged, and immunocompromised SARS-CoV-2 patients who have had secondary fungal infections (SFIs).
Collapse
Affiliation(s)
- Raj Singh
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Mukesh Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Raman Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Md Shamshir Alam
- Department of Pharmacy Practice, College of Pharmacy, National University of Science and Technology, PO Box 620, 130, Bosher-Muscat, Sultanate of Oman
| | - Tapan Kumar Mukherjee
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, UP, India.
- Department of Biotechnology, Amity University, Major Arterial Road, Action Area II, Rajarhat, New Town, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
6
|
Wulandari S, Hartono, Wibawa T. The role of HMGB1 in COVID-19-induced cytokine storm and its potential therapeutic targets: A review. Immunology 2023; 169:117-131. [PMID: 36571562 PMCID: PMC9880760 DOI: 10.1111/imm.13623] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
Hyperinflammation characterized by elevated proinflammatory cytokines known as 'cytokine storms' is the major cause of high severity and mortality seen in COVID-19 patients. The pathology behind the cytokine storms is currently unknown. Increased HMGB1 levels in serum/plasma of COVID-19 patients were reported by many studies, which positively correlated with the level of proinflammatory cytokines. Dead cells following SARS-CoV-2 infection might release a large amount of HMGB1 and RNA of SARS-CoV-2 into extracellular space. HMGB1 is a well-known inflammatory mediator. Additionally, extracellular HMGB1 might interact with SARS-CoV-2 RNA because of its high capability to bind with a wide variety of molecules including nucleic acids and could trigger massive proinflammatory immune responses. This review aimed to critically explore the many possible pathways by which HMGB1-SARS-CoV-2 RNA complexes mediate proinflammatory responses in COVID-19. The contribution of these pathways to impair host immune responses against SARS-CoV-2 infection leading to a cytokine storm was also evaluated. Moreover, since blocking the HMGB1-SARS-CoV-2 RNA interaction might have therapeutic value, some of the HMGB1 antagonists have been reviewed. The HMGB1- SARS-CoV-2 RNA complexes might trigger endocytosis via RAGE which is linked to lysosomal rupture, PRRs activation, and pyroptotic death. High levels of the proinflammatory cytokines produced might suppress many immune cells leading to uncontrolled viral infection and cell damage with more HMGB1 released. Altogether these mechanisms might initiate a proinflammatory cycle leading to a cytokine storm. HMGB1 antagonists could be considered to give benefit in alleviating cytokine storms and serve as a potential candidate for COVID-19 therapy.
Collapse
Affiliation(s)
- Sri Wulandari
- Doctorate Program of Medicine and Health Science, Faculty of MedicinePublic Health and Nursing Universitas Gadjah MadaYogyakartaIndonesia
- Department of Physiology, Faculty of MedicineUniversitas Sebelas MaretSurakartaIndonesia
| | - Hartono
- Department of Physiology, Faculty of MedicineUniversitas Sebelas MaretSurakartaIndonesia
| | - Tri Wibawa
- Department of Microbiology, Faculty of MedicinePublic Health and Nursing Universitas Gadjah MadaYogyakartaIndonesia
| |
Collapse
|
7
|
Nakayama EE, Shioda T. SARS-CoV-2 Related Antibody-Dependent Enhancement Phenomena In Vitro and In Vivo. Microorganisms 2023; 11:microorganisms11041015. [PMID: 37110438 PMCID: PMC10145615 DOI: 10.3390/microorganisms11041015] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies produced in the body after infection or vaccination may enhance subsequent viral infections in vitro and in vivo. Although rare, symptoms of viral diseases are also enhanced by ADE following infection or vaccination in vivo. This is thought to be due to the production of antibodies with low neutralizing activity that bind to the virus and facilitate viral entry, or antigen-antibody complexes that cause airway inflammation, or a predominance of T-helper 2 cells among the immune system cells which leads to excessive eosinophilic tissue infiltration. Notably, ADE of infection and ADE of disease are different phenomena that overlap. In this article, we will describe the three types of ADE: (1) Fc receptor (FcR)-dependent ADE of infection in macrophages, (2) FcR-independent ADE of infection in other cells, and (3) FcR-dependent ADE of cytokine production in macrophages. We will describe their relationship to vaccination and natural infection, and discuss the possible involvement of ADE phenomena in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
8
|
Atmeh PA, Gay L, Levasseur A, La Scola B, Olive D, Mezouar S, Gorvel JP, Mege JL. Macrophages and γδ T cells interplay during SARS-CoV-2 variants infection. Front Immunol 2022; 13:1078741. [PMID: 36601113 PMCID: PMC9806226 DOI: 10.3389/fimmu.2022.1078741] [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: 10/24/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The emergence of several SARS-CoV-2 variants during the COVID pandemic has revealed the impact of variant diversity on viral infectivity and host immune responses. While antibodies and CD8 T cells are essential to clear viral infection, the protective role of innate immunity including macrophages has been recognized. The aims of our study were to compare the infectivity of different SARS-CoV-2 variants in monocyte-derived macrophages (MDM) and to assess their activation profiles and the role of ACE2 (Angiotensin-converting enzyme 2), the main SARS-CoV-2 receptor. We also studied the ability of macrophages infected to affect other immune cells such as γδ2 T cells, another partner of innate immune response to viral infections. Results We showed that the SARS-CoV-2 variants α-B.1.1.7 (United Kingdom), β-B.1.351 (South Africa), γ-P.1 (Brazil), δ-B.1.617 (India) and B.1.1.529 (Omicron), infected MDM without replication, the γ-Brazil variant exhibiting increased infectivity for MDM. No clear polarization profile of SARS-CoV-2 variants-infected MDM was observed. The β-B.1.351 (South Africa) variant induced macrophage activation while B.1.1.529 (Omicron) was rather inhibitory. We observed that SARS-CoV-2 variants modulated ACE2 expression in MDM. In particular, the β-B.1.351 (South Africa) variant induced a higher expression of ACE2, related to MDM activation. Finally, all variants were able to activate γδ2 cells among which γ-P.1 (Brazil) and β-B.1.351 (South Africa) variants were the most efficient. Conclusion Our data show that SARS-CoV-2 variants can infect MDM and modulate their activation, which was correlated with the ACE2 expression. They also affect γδ2 T cell activation. The macrophage response to SARS-CoV-2 variants was stereotypical.
Collapse
Affiliation(s)
- Perla Abou Atmeh
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Laetitia Gay
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Anthony Levasseur
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Bernard La Scola
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Daniel Olive
- Institut Paoli-Calmettes; Aix-Marseille Univ, UM105, Centre National de la Recherche Scientifique (CNRS) UMR 7258, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Jean-Pierre Gorvel
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille Luminy (CIML), Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
- Aix-Marseille Univ, Assitance Publique Hopitaux de Marseille (APHM), Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| |
Collapse
|
9
|
Ranjbar M, Rahimi A, Baghernejadan Z, Ghorbani A, Khorramdelazad H. Role of CCL2/CCR2 axis in the pathogenesis of COVID-19 and possible Treatments: All options on the Table. Int Immunopharmacol 2022; 113:109325. [PMID: 36252475 PMCID: PMC9561120 DOI: 10.1016/j.intimp.2022.109325] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is cause of the novel coronavirus disease (COVID-19). In the last two years, SARS-CoV-2 has infected millions of people worldwide with different waves, resulting in the death of many individuals. The evidence disclosed that the host immune responses to SARS-CoV-2 play a pivotal role in COVID-19 pathogenesis and clinical manifestations. In addition to inducing antiviral immune responses, SARS-CoV-2 can also cause dysregulated inflammatory responses characterized by the noticeable release of proinflammatory mediators in COVID-19 patients. Among these proinflammatory mediators, chemokines are considered a subset of cytokines that participate in the chemotaxis process to recruit immune and non-immune cells to the site of inflammation and infection. Researchers have demonstrated that monocyte chemoattractant protein-1 (MCP-1/CCL2) and its receptor (CCR2) are involved in the recruitment of monocytes and infiltration of these cells into the lungs of patients suffering from COVID-19. Moreover, elevated levels of CCL2 have been reported in the bronchoalveolar lavage fluid (BALF) obtained from patients with severe COVID-19, initiating cytokine storm and promoting CD163+ myeloid cells infiltration in the airways and further alveolar damage. Therefore, CCL2/CCR axis plays a key role in the immunopathogenesis of COVID-19 and targeted therapy of involved molecules in this axis can be a potential therapeutic approach for these patients. This review discusses the biology of the CCL2/CCR2 axis as well as the role of this axis in COVID-19 immunopathogenesis, along with therapeutic options aimed at inhibiting CCL2/CCR2 and modulating dysregulated inflammatory responses in patients with severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mitra Ranjbar
- Department of Infectious Disease, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atousa Ghorbani
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
10
|
Mi Y, Liang L, Xu K, Li Q, Wang W, Dang W, Deng J, Zhi Y, Li X, Tan J. Severe acute respiratory syndrome coronavirus 2 virus-like particles induce dendritic cell maturation and modulate T cell immunity. Front Cell Infect Microbiol 2022; 12:986350. [DOI: 10.3389/fcimb.2022.986350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that play an important role in both innate and acquired immune responses against pathogens. However, the role of DCs in coronavirus disease 2019 (COVID-19) is unclear. Virus-like particles (VLPs) that structurally mimic the original virus are one of the candidates COVID-19 vaccines. In the present study, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VLPs were used as an alternative to live virus to evaluate the interaction of the virus with DCs. The results revealed that SARS-CoV-2 VLPs induced DC maturation by augmenting cell surface molecule expression (CD80, CD86, and major histocompatibility complex class II (MHC-II)) and inflammatory cytokine production (tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-12p70) in DCs via the mitogen-activated protein kinase and nuclear factor-κB signaling pathways. In addition, mature DCs induced by SARS-CoV-2 VLPs promoted T cell proliferation, which was dependent on VLPs concentration. Our results suggest that SARS-CoV-2 VLPs regulate the immune response by interacting with DCs. These findings will improve the understanding of SARS-CoV-2 pathogenesis and SARS-CoV-2 vaccine development.
Collapse
|
11
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
12
|
Kazmierski J, Friedmann K, Postmus D, Emanuel J, Fischer C, Jansen J, Richter A, Bosquillon de Jarcy L, Schüler C, Sohn M, Sauer S, Drosten C, Saliba A, Sander LE, Müller MA, Niemeyer D, Goffinet C. Nonproductive exposure of PBMCs to SARS-CoV-2 induces cell-intrinsic innate immune responses. Mol Syst Biol 2022; 18:e10961. [PMID: 35975552 PMCID: PMC9382356 DOI: 10.15252/msb.202210961] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Cell-intrinsic responses mounted in PBMCs during mild and severe COVID-19 differ quantitatively and qualitatively. Whether they are triggered by signals emitted by productively infected cells of the respiratory tract or result from physical interaction with virus particles remains unclear. Here, we analyzed susceptibility and expression profiles of PBMCs from healthy donors upon ex vivo exposure to SARS-CoV and SARS-CoV-2. In line with the absence of detectable ACE2 receptor expression, human PBMCs were refractory to productive infection. RT-PCR experiments and single-cell RNA sequencing revealed JAK/STAT-dependent induction of interferon-stimulated genes (ISGs) but not proinflammatory cytokines. This SARS-CoV-2-specific response was most pronounced in monocytes. SARS-CoV-2-RNA-positive monocytes displayed a lower ISG signature as compared to bystander cells of the identical culture. This suggests a preferential invasion of cells with a low ISG baseline profile or delivery of a SARS-CoV-2-specific sensing antagonist upon efficient particle internalization. Together, nonproductive physical interaction of PBMCs with SARS-CoV-2- and, to a much lesser extent, SARS-CoV particles stimulate JAK/STAT-dependent, monocyte-accentuated innate immune responses that resemble those detected in vivo in patients with mild COVID-19.
Collapse
Affiliation(s)
- Julia Kazmierski
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Kirstin Friedmann
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Jackson Emanuel
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Cornelius Fischer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Anja Richter
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Laure Bosquillon de Jarcy
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Christiane Schüler
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Madlen Sohn
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Sascha Sauer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Antoine‐Emmanuel Saliba
- Helmholtz Institute for RNA‐based Infection Research (HIRI)Helmholtz‐Center for Infection Research (HZI)WürzburgGermany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Marcel A Müller
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| |
Collapse
|
13
|
Ortega-Bernal D, Zarate S, Martinez-Cárdenas MDLÁ, Bojalil R. An approach to cellular tropism of SARS-CoV-2 through protein-protein interaction and enrichment analysis. Sci Rep 2022; 12:9399. [PMID: 35672403 PMCID: PMC9172986 DOI: 10.1038/s41598-022-13625-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 05/17/2022] [Indexed: 01/17/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, is a primarily pulmonary disease that can affect several organs, directly or indirectly. To date, there are many questions about the different pathological mechanisms. Here, we generate an approach to identify the cellular-level tropism of SARS-CoV-2 using human proteomics, virus-host interactions, and enrichment analysis. Through a network-based approach, the molecular context was visualized and analyzed. This procedure was also performed for SARS-CoV-1. We obtained proteomes and interactomes from 145 different cells corresponding to 57 different tissues. We discarded the cells without the proteins known for interacting with the virus, such as ACE2 or TMPRSS2. Of the remaining cells, a gradient of susceptibility to infection was observed. In addition, we identified proteins associated with the coagulation cascade that can be directly or indirectly affected by viral proteins. As a whole we identified 55 cells that could be potentially controlled by the virus, with different susceptibilities, mainly being pneumocytes, heart, kidney, liver, or small intestine cells. These results help to explain the molecular context and provide elements for possible treatments in the current situation. This strategy may be useful for other viruses, especially those with limited reported PPI, such as a new virus.
Collapse
Affiliation(s)
- Daniel Ortega-Bernal
- Department of Health Care, Universidad Autónoma Metropolitana, Unidad Xochimilco, 04960, Mexico City, Mexico
| | - Selene Zarate
- Posgrado en Ciencias Genómicas, Universidad Autónoma de La Ciudad de México, Ciudad de México, Mexico City, 03100, México
| | | | - Rafael Bojalil
- Department of Health Care, Universidad Autónoma Metropolitana, Unidad Xochimilco, 04960, Mexico City, Mexico.
| |
Collapse
|
14
|
Nakayama EE, Kubota-Koketsu R, Sasaki T, Suzuki K, Uno K, Shimizu J, Okamoto T, Matsumoto H, Matsuura H, Hashimoto S, Tanaka T, Harada H, Tomita M, Kaneko M, Yoshizaki K, Shioda T. Anti-nucleocapsid antibodies enhance the production of IL-6 induced by SARS-CoV-2 N protein. Sci Rep 2022; 12:8108. [PMID: 35577892 PMCID: PMC9109953 DOI: 10.1038/s41598-022-12252-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022] Open
Abstract
A cytokine storm induces acute respiratory distress syndrome, the main cause of death in coronavirus disease 2019 (COVID-19) patients. However, the detailed mechanisms of cytokine induction due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain unclear. To examine the cytokine production in COVID-19, we mimicked the disease in SARS-CoV-2-infected alveoli by adding the lysate of SARS-CoV-2-infected cells to cultured macrophages or induced pluripotent stem cell-derived myeloid cells. The cells secreted interleukin (IL)-6 after the addition of SARS-CoV-2-infected cell lysate. Screening of 25 SARS-CoV-2 protein-expressing plasmids revealed that the N protein-coding plasmid alone induced IL-6 production. The addition of anti-N antibody further enhanced IL-6 production, but the F(ab')2 fragment did not. Sera from COVID-19 patients also enhanced IL-6 production, and sera from patients with severer disease induced higher levels of IL-6. These results suggest that anti-N antibody promotes IL-6 production in SARS-CoV-2-infected alveoli, leading to the cytokine storm of COVID-19.
Collapse
Affiliation(s)
- Emi E Nakayama
- Research Institute for Microbial Diseases and Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Ritsuko Kubota-Koketsu
- Research Institute for Microbial Diseases and Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tadahiro Sasaki
- Research Institute for Microbial Diseases and Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Keita Suzuki
- Research Institute for Microbial Diseases and Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan.,TANAKA Kikinzoku Kogyo K.K, Hiratsuka, Kanagawa, 254-0076, Japan
| | - Kazuko Uno
- Division of Basic Research, Louis Pasteur Center for Medical Research, Kyoto, 606-8225, Japan
| | - Jun Shimizu
- MiCAN Technologies Inc., Kyoto, 615-8245, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0781, Japan
| | - Hisatake Matsumoto
- Trauma and Acute Critical Care Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Matsuura
- Osaka Prefectural Nakakawachi Emergency and Critical Care Center, Higashiosaka, Osaka, 678-0947, Japan
| | - Shoji Hashimoto
- Osaka Prefectural Hospital Organization Osaka Habikino Medical Center, Habikino, Osaka, 583-8588, Japan
| | - Toshio Tanaka
- Osaka Prefectural Hospital Organization Osaka Habikino Medical Center, Habikino, Osaka, 583-8588, Japan
| | - Hiromasa Harada
- Yao Tokushukai General Hospital, Yao, Osaka, 581-0011, Japan
| | | | | | - Kazuyuki Yoshizaki
- Institute of Scientific and Industry Research, Osaka University, Ibaraki, Osaka, 567-0047, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases and Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
15
|
Dhaliwal M, Tyagi R, Malhotra P, Barman P, Loganathan SK, Sharma J, Sharma K, Mondal S, Rawat A, Singh S. Mechanisms of Immune Dysregulation in COVID-19 Are Different From SARS and MERS: A Perspective in Context of Kawasaki Disease and MIS-C. Front Pediatr 2022; 10:790273. [PMID: 35601440 PMCID: PMC9119432 DOI: 10.3389/fped.2022.790273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Coronaviruses have led to three major outbreaks to date-Severe Acute Respiratory Syndrome (SARS; 2002), Middle East Respiratory Syndrome (MERS; 2012) and the ongoing pandemic, Coronavirus Disease (COVID-19; 2019). Coronavirus infections are usually mild in children. However, a few children with MERS had presented with a severe phenotype in the acute phase resulting in progressive pneumonic changes with increasing oxygen dependency and acute respiratory distress requiring ventilatory support. A subset of children with a history of SARS-CoV-2 infection develops a multisystem hyper-inflammatory phenotype known as Multisystem Inflammatory Syndrome in Children (MIS-C). This syndrome occurs 4-6 weeks after infection with SARS-CoV-2 and has been reported more often from areas with high community transmission. Children with MIS-C present with high fever and often have involvement of cardiovascular, gastrointestinal and hematologic systems leading to multiorgan failure. This is accompanied by elevation of pro-inflammatory cytokines such as IL-6 and IL-10. MIS-C has several similarities with Kawasaki disease (KD) considering children with both conditions present with fever, rash, conjunctival injection, mucosal symptoms and swelling of hands and feet. For reasons that are still not clear, both KD and MIS-C were not reported during the SARS-CoV and MERS-CoV outbreaks. As SARS-CoV-2 differs from SARS-CoV by 19.5% and MERS by 50% in terms of sequence identity, differences in genomic and proteomic profiles may explain the varied disease immunopathology and host responses. Left untreated, MIS-C may lead to severe abdominal pain, ventricular dysfunction and shock. Immunological investigations reveal reduced numbers of follicular B cells, increased numbers of terminally differentiated CD4+T lymphocytes, and decreased IL-17A. There is still ambiguity about the clinical and immunologic risk factors that predispose some children to development of MIS-C while sparing others. Host-pathogen interactions in SARS, MERS and COVID-19 are likely to play a crucial role in the clinical phenotypes that manifest. This narrative review focuses on the immunological basis for development of MIS-C syndrome in the ongoing SARS-CoV-2 pandemic. To the best of our knowledge, these aspects have not been reviewed before.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Surjit Singh
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
16
|
Gao J, Mei H, Sun J, Li H, Huang Y, Tang Y, Duan L, Liu D, Pang Y, Wang Q, Gao Y, Song K, Zhao J, Zhang C, Liu J. Neuropilin-1-Mediated SARS-CoV-2 Infection in Bone Marrow-Derived Macrophages Inhibits Osteoclast Differentiation. Adv Biol (Weinh) 2022; 6:e2200007. [PMID: 35195371 PMCID: PMC9073998 DOI: 10.1002/adbi.202200007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/25/2022] [Indexed: 01/27/2023]
Abstract
In humans, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can cause medical complications across various tissues and organs. Despite the advances to understanding the pathogenesis of SARS-CoV-2, its tissue tropism and interactions with host cells have not been fully understood. Existing clinical data have revealed disordered calcium and phosphorus metabolism in Coronavirus Disease 2019 (COVID-19) patients, suggesting possible infection or damage in the human skeleton system by SARS-CoV-2. Herein, SARS-CoV-2 infection in mouse models with wild-type and beta strain (B.1.351) viruses is investigated, and it is found that bone marrow-derived macrophages (BMMs) can be efficiently infected in vivo. Single-cell RNA sequencing (scRNA-Seq) analyses of infected BMMs identify distinct clusters of susceptible macrophages, including those related to osteoblast differentiation. Interestingly, SARS-CoV-2 entry on BMMs is dependent on the expression of neuropilin-1 (NRP1) rather than the widely recognized receptor angiotensin-converting enzyme 2 (ACE2). The loss of NRP1 expression during BMM-to-osteoclast differentiation or NRP1 neutralization and knockdown can significantly inhibit SARS-CoV-2 infection in BMMs. Importantly, it is found that authentic SARS-CoV-2 infection impedes BMM-to-osteoclast differentiation. Collectively, this study provides evidence for NRP1-mediated SARS-CoV-2 infection in BMMs and establishes a potential link between disturbed osteoclast differentiation and disordered skeleton metabolism in COVID-19 patients.
Collapse
Affiliation(s)
- Junjie Gao
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Jing Sun
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510182China
| | - Hao Li
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Yuege Huang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China,Shanghai Clinical Research and Trial CenterShanghai201210China,Gene Editing CenterSchool of Life Science and TechnologyShanghaiTech UniversityShanghai201210China,University of Chinese Academy of SciencesBeijing100049China
| | - Yanhong Tang
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510182China
| | - Linwei Duan
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510182China
| | - Delin Liu
- Centre for Orthopaedic ResearchSchool of SurgeryThe University of Western AustraliaNedlandsWestern Australia6009Australia
| | - Yidan Pang
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Qiyang Wang
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Youshui Gao
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Ke Song
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Jincun Zhao
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510182China
| | - Changqing Zhang
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Shanghai Sixth People's HospitalShanghai200233China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China,Shanghai Clinical Research and Trial CenterShanghai201210China,Gene Editing CenterSchool of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| |
Collapse
|
17
|
Pontelli MC, Castro IA, Martins RB, La Serra L, Veras FP, Nascimento DC, Silva CM, Cardoso RS, Rosales R, Gomes R, Lima TM, Souza JP, Vitti BC, Caetité DB, de Lima MHF, Stumpf SD, Thompson CE, Bloyet LM, Kawahisa JTE, Giannini MC, Bonjorno LP, Lopes MIF, Batah SS, Li S, Assad RL, Almeida SCL, Oliveira FR, Benatti MN, Pontes LLF, Santana RC, Vilar FC, Martins MA, Shi PY, Cunha TM, Calado RT, Alves-Filho JC, Zamboni DS, Fabro A, Louzada-Junior P, Oliveira RDR, Whelan SPJ, Cunha FQ, Arruda E. SARS-CoV-2 productively infects primary human immune system cells in vitro and in COVID-19 patients. J Mol Cell Biol 2022; 14:6572370. [PMID: 35451490 PMCID: PMC9384834 DOI: 10.1093/jmcb/mjac021] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/30/2021] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with a hyperinflammatory state and lymphocytopenia, a hallmark that appears as both signature and prognosis of disease severity outcome. Although cytokine storm and a sustained inflammatory state are commonly associated with immune cell depletion, it is still unclear whether direct SARS-CoV-2 infection of immune cells could also play a role in this scenario by harboring viral replication. We found that monocytes, as well as both B and T lymphocytes, were susceptible to SARS-CoV-2 infection in vitro, accumulating double-stranded RNA consistent with viral RNA replication and ultimately leading to expressive T cell apoptosis. In addition, flow cytometry and immunofluorescence analysis revealed that SARS-CoV-2 was frequently detected in monocytes and B lymphocytes from coronavirus disease 2019 (COVID-19) patients. The rates of SARS-CoV-2-infected monocytes in peripheral blood mononuclear cells from COVID-19 patients increased over time from symptom onset, with SARS-CoV-2-positive monocytes, B cells, and CD4+ T lymphocytes also detected in postmortem lung tissue. These results indicated that SARS-CoV-2 infection of blood-circulating leukocytes in COVID-19 patients might have important implications for disease pathogenesis and progression, immune dysfunction, and virus spread within the host.
Collapse
Affiliation(s)
- Marjorie C Pontelli
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Italo A Castro
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Ronaldo B Martins
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Leonardo La Serra
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Flávio P Veras
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Daniele C Nascimento
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Camila M Silva
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Ricardo S Cardoso
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Roberta Rosales
- Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Rogério Gomes
- Blood Center of Ribeirao Preto, 14049-900, Ribeirao Preto, São Paulo, Brazil
| | - Thais M Lima
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Juliano P Souza
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Brenda C Vitti
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Diego B Caetité
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Mikhael H F de Lima
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Spencer D Stumpf
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cassandra E Thompson
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Louis-Marie Bloyet
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Juliana T E Kawahisa
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Marcela C Giannini
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Letícia P Bonjorno
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Maria I F Lopes
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Sabrina S Batah
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Siyuan Li
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Rodrigo L Assad
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Sergio C L Almeida
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Fabiola R Oliveira
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Maíra N Benatti
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Lorena L F Pontes
- Blood Center of Ribeirao Preto, 14049-900, Ribeirao Preto, São Paulo, Brazil
| | - Rodrigo C Santana
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando C Vilar
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Maria A Martins
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Rodrigo T Calado
- Blood Center of Ribeirao Preto, 14049-900, Ribeirao Preto, São Paulo, Brazil
| | - José C Alves-Filho
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Dario S Zamboni
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Alexandre Fabro
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Paulo Louzada-Junior
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Rene D R Oliveira
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Divisions of Clinical Immunology, Infectious Diseases and Intensive Care Unit, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Fernando Q Cunha
- Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| | - Eurico Arruda
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil.,Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of Sao Paulo, 14049-900, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
18
|
Niedźwiedzka-Rystwej P, Majchrzak A, Kurkowska S, Małkowska P, Sierawska O, Hrynkiewicz R, Parczewski M. Immune Signature of COVID-19: In-Depth Reasons and Consequences of the Cytokine Storm. Int J Mol Sci 2022; 23:4545. [PMID: 35562935 PMCID: PMC9105989 DOI: 10.3390/ijms23094545] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
In the beginning of the third year of the fight against COVID-19, the virus remains at least still one step ahead in the pandemic "war". The key reasons are evolving lineages and mutations, resulting in an increase of transmissibility and ability to evade immune system. However, from the immunologic point of view, the cytokine storm (CS) remains a poorly understood and difficult to combat culprit of the extended number of in-hospital admissions and deaths. It is not fully clear whether the cytokine release is a harmful result of suppression of the immune system or a positive reaction necessary to clear the virus. To develop methods of appropriate treatment and therefore decrease the mortality of the so-called COVID-19-CS, we need to look deeply inside its pathogenesis, which is the purpose of this review.
Collapse
Affiliation(s)
| | - Adam Majchrzak
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| | - Sara Kurkowska
- Department of Nuclear Medicine, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Paulina Małkowska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Olga Sierawska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| |
Collapse
|
19
|
Ekanger CT, Zhou F, Bohan D, Lotsberg ML, Ramnefjell M, Hoareau L, Røsland GV, Lu N, Aanerud M, Gärtner F, Salminen PR, Bentsen M, Halvorsen T, Ræder H, Akslen LA, Langeland N, Cox R, Maury W, Stuhr LEB, Lorens JB, Engelsen AST. Human Organotypic Airway and Lung Organoid Cells of Bronchiolar and Alveolar Differentiation Are Permissive to Infection by Influenza and SARS-CoV-2 Respiratory Virus. Front Cell Infect Microbiol 2022; 12:841447. [PMID: 35360113 PMCID: PMC8964279 DOI: 10.3389/fcimb.2022.841447] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has led to the initiation of unprecedented research efforts to understand the pathogenesis mediated by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). More knowledge is needed regarding the cell type-specific cytopathology and its impact on cellular tropism. Furthermore, the impact of novel SARS-CoV-2 mutations on cellular tropism, alternative routes of entry, the impact of co-infections, and virus replication kinetics along the respiratory tract remains to be explored in improved models. Most applied virology models are not well suited to address the remaining questions, as they do not recapitulate the histoarchitecture and cellular composition of human respiratory tissues. The overall aim of this work was to establish from single biopsy specimens, a human adult stem cell-derived organoid model representing the upper respiratory airways and lungs and explore the applicability of this model to study respiratory virus infection. First, we characterized the organoid model with respect to growth pattern and histoarchitecture, cellular composition, and functional characteristics. Next, in situ expression of viral entry receptors, including influenza virus-relevant sialic acids and SARS-CoV-2 entry receptor ACE2 and TMPRSS2, were confirmed in organoids of bronchiolar and alveolar differentiation. We further showed successful infection by pseudotype influenza A H7N1 and H5N1 virus, and the ability of the model to support viral replication of influenza A H7N1 virus. Finally, successful infection and replication of a clinical isolate of SARS-CoV-2 were confirmed in the organoids by TCID50 assay and immunostaining to detect intracellular SARS-CoV-2 specific nucleocapsid and dsRNA. The prominent syncytia formation in organoid tissues following SARS-CoV-2 infection mimics the findings from infected human tissues in situ. We conclude that the human organotypic model described here may be particularly useful for virology studies to evaluate regional differences in the host response to infection. The model contains the various cell types along the respiratory tract, expresses respiratory virus entry factors, and supports successful infection and replication of influenza virus and SARS-CoV-2. Thus, the model may serve as a relevant and reliable tool in virology and aid in pandemic preparedness, and efficient evaluation of antiviral strategies.
Collapse
Affiliation(s)
- Camilla Tvedt Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Fan Zhou
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Maria Lie Lotsberg
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Maria Ramnefjell
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Laurence Hoareau
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Gro Vatne Røsland
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Ning Lu
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Marianne Aanerud
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Fabian Gärtner
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Pirjo Riitta Salminen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Section of Cardiothoracic Surgery, Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Mariann Bentsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Thomas Halvorsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Helge Ræder
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Nina Langeland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Cox
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | | | - James B. Lorens
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- *Correspondence: Agnete S. T. Engelsen,
| |
Collapse
|
20
|
Natarajan H, Xu S, Crowley AR, Butler SE, Weiner JA, Bloch EM, Littlefield K, Benner SE, Shrestha R, Ajayi O, Wieland-Alter W, Sullivan D, Shoham S, Quinn TC, Casadevall A, Pekosz A, Redd AD, Tobian AAR, Connor RI, Wright PF, Ackerman ME. Antibody attributes that predict the neutralization and effector function of polyclonal responses to SARS-CoV-2. BMC Immunol 2022; 23:7. [PMID: 35172720 PMCID: PMC8851712 DOI: 10.1186/s12865-022-00480-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/07/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND While antibodies can provide significant protection from SARS-CoV-2 infection and disease sequelae, the specific attributes of the humoral response that contribute to immunity are incompletely defined. METHODS We employ machine learning to relate characteristics of the polyclonal antibody response raised by natural infection to diverse antibody effector functions and neutralization potency with the goal of generating both accurate predictions of each activity based on antibody response profiles as well as insights into antibody mechanisms of action. RESULTS To this end, antibody-mediated phagocytosis, cytotoxicity, complement deposition, and neutralization were accurately predicted from biophysical antibody profiles in both discovery and validation cohorts. These models identified SARS-CoV-2-specific IgM as a key predictor of neutralization activity whose mechanistic relevance was supported experimentally by depletion. CONCLUSIONS Validated models of how different aspects of the humoral response relate to antiviral antibody activities suggest desirable attributes to recapitulate by vaccination or other antibody-based interventions.
Collapse
Affiliation(s)
- Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Shiwei Xu
- Program in Quantitative Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Savannah E Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah E Benner
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ruchee Shrestha
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Olivia Ajayi
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Wendy Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - David Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas C Quinn
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ruth I Connor
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
- Program in Quantitative Biological Sciences, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.
| |
Collapse
|
21
|
Abstract
The process of adaptation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to humans probably had started decades ago, when its ancestor diverged from the bat coronavirus. The adaptive process comprises strategies the virus uses to overcome the respiratory tract defense barriers and replicate and shed in the host cells. These strategies include the impairment of interferon production, hiding immunogenic motifs, avoiding viral RNA detection, manipulating cell autophagy, triggering host cell death, inducing lymphocyte exhaustion and depletion, and finally, mutation and escape from immunity. In addition, SARS-CoV-2 employs strategies to take advantage of host cell resources for its benefits, such as inhibiting the ubiquitin-proteasome system, hijacking mitochondria functions, and usage of enhancing antibodies. It may be anticipated that as the tradeoffs of adaptation progress, the virus destructive burden will gradually subside. Some evidence suggests that SARS-CoV-2 will become part of the human respiratory virome, as had occurred with other coronaviruses, and coevolve with its host.
Collapse
Affiliation(s)
- Eduardo Tosta
- Universidade de Brasília, Faculdade de Medicina, Brasília, DF, Brasil
| |
Collapse
|
22
|
Kumar IC, Babu M, Tripathi M. A study to identify severe acute respiratory syndrome coronavirus 2 in erythrocytes of patients suffering from coronavirus disease-19 at an Apex tertiary care institute in Andhra Pradesh, South India. Asian J Transfus Sci 2022. [DOI: 10.4103/ajts.ajts_98_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Uddin MB, Sajib EH, Hoque SF, Hassan MM, Ahmed SSU. Macrophages in respiratory system. RECENT ADVANCEMENTS IN MICROBIAL DIVERSITY 2022:299-333. [DOI: 10.1016/b978-0-12-822368-0.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
24
|
Veenhuis RT, Zeiss CJ. Animal Models of COVID-19 II. Comparative Immunology. ILAR J 2021; 62:17-34. [PMID: 33914873 PMCID: PMC8135340 DOI: 10.1093/ilar/ilab010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022] Open
Abstract
Developing strong animal models is essential for furthering our understanding of how the immune system functions in response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. The alarming speed at which SARS-CoV-2 has spread, and the high mortality rate of severe Coronavirus Disease 2019 (COVID-19), has required both basic science and clinical research to move at an unprecedented pace. Models previously developed to study the immune response against SARS-CoV have been rapidly deployed to now study SARS-CoV-2. To date, both small and large animal models are remarkably consistent when infected with SARS-CoV-2; however, certain models have proven more useful when answering specific immunological questions than others. Small animal models, such as Syrian hamsters, ferrets, and mice carrying the hACE2 transgene, appear to reliably recapitulate the initial cytokine surge seen in COVID-19 as well as show significant innate and adaptive cell infiltration in to the lung early in infection. Additionally, these models develop strong antibody responses to the virus, are protected from reinfection, and genetically modified versions exist that can be used to ask specific immunological questions. Large animal models such as rhesus and cynomologus macaques and African green monkeys are critical to understanding how the immune system responds to SARS-CoV-2 infection because they are considered to be the most similar to humans. These models are considered the gold standard for assessing vaccine efficacy and protection, and recapitulate the initial cytokine surge, immune cell infiltration into the lung, certain aspects of thrombosis, and the antibody and T-cell response to the virus. In this review, we discuss both small and large animal model studies previously used in SARS-CoV-2 research that may be useful in elucidating the immunological contributions to hallmark syndromes observed with COVID-19.
Collapse
Affiliation(s)
- Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Almutairi MM, Sivandzade F, Albekairi TH, Alqahtani F, Cucullo L. Neuroinflammation and Its Impact on the Pathogenesis of COVID-19. Front Med (Lausanne) 2021; 8:745789. [PMID: 34901061 PMCID: PMC8652056 DOI: 10.3389/fmed.2021.745789] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The clinical manifestations of COVID-19 include dry cough, difficult breathing, fever, fatigue, and may lead to pneumonia and respiratory failure. There are significant gaps in the current understanding of whether SARS-CoV-2 attacks the CNS directly or through activation of the peripheral immune system and immune cell infiltration. Although the modality of neurological impairments associated with COVID-19 has not been thoroughly investigated, the latest studies have observed that SARS-CoV-2 induces neuroinflammation and may have severe long-term consequences. Here we review the literature on possible cellular and molecular mechanisms of SARS-CoV-2 induced-neuroinflammation. Activation of the innate immune system is associated with increased cytokine levels, chemokines, and free radicals in the SARS-CoV-2-induced pathogenic response at the blood-brain barrier (BBB). BBB disruption allows immune/inflammatory cell infiltration into the CNS activating immune resident cells (such as microglia and astrocytes). This review highlights the molecular and cellular mechanisms involved in COVID-19-induced neuroinflammation, which may lead to neuronal death. A better understanding of these mechanisms will help gain substantial knowledge about the potential role of SARS-CoV-2 in neurological changes and plan possible therapeutic intervention strategies.
Collapse
Affiliation(s)
- Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Farzane Sivandzade
- Department of Biological Sciences, Oakland University, Rochester, MI, United States
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Luca Cucullo
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| |
Collapse
|
26
|
Baradaran Ghavami S, Pourhamzeh M, Farmani M, Keshavarz H, Shahrokh S, Shpichka A, Asadzadeh Aghdaei H, Hakemi-Vala M, Hossein-khannazer N, Timashev P, Vosough M. Cross-talk between immune system and microbiota in COVID-19. Expert Rev Gastroenterol Hepatol 2021; 15:1281-1294. [PMID: 34654347 PMCID: PMC8567289 DOI: 10.1080/17474124.2021.1991311] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/06/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human gut microbiota plays a crucial role in providing protective responses against pathogens, particularly by regulating immune system homeostasis. There is a reciprocal interaction between the gut and lung microbiota, called the gut-lung axis (GLA). Any alteration in the gut microbiota or their metabolites can cause immune dysregulation, which can impair the antiviral activity of the immune system against respiratory viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. AREAS COVERED This narrative review mainly outlines emerging data on the mechanisms underlying the interactions between the immune system and intestinal microbial dysbiosis, which is caused by an imbalance in the levels of essential metabolites. The authors will also discuss the role of probiotics in restoring the balance of the gut microbiota and modulation of cytokine storm. EXPERT OPINION Microbiota-derived signals regulate the immune system and protect different tissues during severe viral respiratory infections. The GLA's equilibration could help manage the mortality and morbidity rates associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Hediye Keshavarz
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Shabnam Shahrokh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mojdeh Hakemi-Vala
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-khannazer
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
27
|
von Massow G, Oh S, Lam A, Gustafsson K. Gamma Delta T Cells and Their Involvement in COVID-19 Virus Infections. Front Immunol 2021; 12:741218. [PMID: 34777353 PMCID: PMC8586491 DOI: 10.3389/fimmu.2021.741218] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/05/2021] [Indexed: 12/29/2022] Open
Abstract
The global outbreak of the SARS-Cov-2 virus in 2020 has killed millions of people worldwide and forced large parts of the world into lockdowns. While multiple vaccine programs are starting to immunize the global population, there is no direct cure for COVID-19, the disease caused by the SARS-Cov-2 infection. A common symptom in patients is a decrease in T cells, called lymphopenia. It is as of yet unclear what the exact role of T cells are in the immune response to COVID-19. The research so far has mainly focused on the involvement of classical αβ T cells. However, another subset of T cells called γδ T cells could have an important role to play. As part of the innate immune system, γδ T cells respond to inflammation and stressed or infected cells. The γδ T cell subset appears to be particularly affected by lymphopenia in COVID-19 patients and commonly express activation and exhaustion markers. Particularly in children, this subset of T cells seems to be most affected. This is interesting and relevant because γδ T cells are more prominent and active in early life. Their specific involvement in this group of patients could indicate a significant role for γδ T cells in this disease. Furthermore, they seem to be involved in other viral infections and were able to kill SARS infected cells in vitro. γδ T cells can take up, process and present antigens from microbes and human cells. As e.g. tumour-associated antigens are presented by MHC on γδ T cells to classical T-cells, we argue here that it stands to reason that also viral antigens, such as SARS-Cov-2-derived peptides, can be presented in the same way. γδ T cells are already used for medical purposes in oncology and have potential in cancer therapy. As γδ T cells are not necessarily able to distinguish between a transformed and a virally infected cell it could therefore be of great interest to investigate further the relationship between COVID-19 and γδ T cells.
Collapse
Affiliation(s)
- Georg von Massow
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore
| | - Alan Lam
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore
| | - Kenth Gustafsson
- Department of Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
28
|
Macrophages and Monocytes: "Trojan Horses" in COVID-19. Viruses 2021; 13:v13112178. [PMID: 34834984 PMCID: PMC8624282 DOI: 10.3390/v13112178] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/25/2021] [Indexed: 01/18/2023] Open
Abstract
We aimed to explore whether variants of SARS-CoV-2 (Chinese-derived strain (D614, lineage A), Italian strain PV10734 (D614G, lineage B.1.1) and Alpha strain (lineage B.1.1.7)) were able to infect monocytes (MN) and monocyte-derived macrophages (MDM) and whether these infected cells may, in turn, be vectors of infection. For this purpose, we designed an in vitro study following the evolution of MN and MDM infection at different time points in order to confirm whether these cells were permissive for SARS-CoV-2 replication. Finally, we investigated whether, regardless of viral replication, the persistent virus can be transferred to non-infected cells permissive for viral replication. Thus, we co-cultured the infected MN/MDM with permissive VERO E6 cells verifying the viral transmission. This is a further in vitro demonstration of the important role of MN and MDM in the dissemination of SARS-CoV-2 and evolution of the COVID-19 disease.
Collapse
|
29
|
Agrawal S, Salazar J, Tran TM, Agrawal A. Sex-Related Differences in Innate and Adaptive Immune Responses to SARS-CoV-2. Front Immunol 2021; 12:739757. [PMID: 34745109 PMCID: PMC8563790 DOI: 10.3389/fimmu.2021.739757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/28/2021] [Indexed: 01/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) exhibits a sex bias with males showing signs of more severe disease and hospitalizations compared with females. The mechanisms are not clear but differential immune responses, particularly the initial innate immune response, between sexes may be playing a role. The early innate immune responses to SARS-CoV-2 have not been studied because of the gap in timing between the patient becoming infected, showing symptoms, and getting the treatment. The primary objective of the present study was to compare the response of dendritic cells (DCs) and monocytes from males and females to SARS-CoV-2, 24 h after infection. To investigate this, peripheral blood mononuclear cells (PBMCs) from healthy young individuals were stimulated in vitro with the virus. Our results indicate that PBMCs from females upregulated the expression of HLA-DR and CD86 on pDCs and mDCs after stimulation with the virus, while the activation of these cells was not significant in males. Monocytes from females also displayed increased activation than males. In addition, females secreted significantly higher levels of IFN-α and IL-29 compared with males at 24 h. However, the situation was reversed at 1 week post stimulation and males displayed high levels of IFN-α production compared with females. Further investigations revealed that the secretion of CXCL-10, a chemokine associated with lung complications, was higher in males than females at 24 h. The PBMCs from females also displayed increased induction of CTLs. Altogether, our results suggest that decreased activation of pDCs, mDCs, and monocytes and the delayed and prolonged IFN-α secretion along with increased CXCL-10 secretion may be responsible for the increased morbidity and mortality of males to COVID-19.
Collapse
Affiliation(s)
| | | | | | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
30
|
Palladino M. Complete blood count alterations in COVID-19 patients: A narrative review. Biochem Med (Zagreb) 2021; 31:030501. [PMID: 34658642 PMCID: PMC8495616 DOI: 10.11613/bm.2021.030501] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic represents a scientific and social crisis. One of the main unmet needs for coronavirus disease 2019 is its unpredictable clinical course, which can rapidly change in an irreversible outcome. COVID-19 patients can be classified into mild, moderate, and severe. Several haematological parameters, such as platelets, white blood cell total count, lymphocytes, neutrophils, (together with neutrophil-lymphocyte and platelet-lymphocyte ratio), and haemoglobin were described to be associated with COVID-19 infection and severity. The purpose of these review is to describe the current state of the art about complete blood count alterations during COVID-19 infection, and to summarize the crucial role of some haematological parameters during the course of the disease. Decreased platelet, lymphocyte, haemoglobin, eosinophil, and basophil count, increased neutrophil count and neutrophil-lymphocyte and platelet-lymphocyte ratio have been associated with COVID-19 infection and a worse clinical outcome. Our study adds some novelty about the identification of effective biomarkers of progressive disease, and might be helpful for diagnosis, prevention of complications, and effective therapy.
Collapse
|
31
|
Abdelmoaty MM, Yeapuri P, Machhi J, Olson KE, Shahjin F, Kumar V, Zhou Y, Liang J, Pandey K, Acharya A, Byrareddy SN, Mosley RL, Gendelman HE. Defining the Innate Immune Responses for SARS-CoV-2-Human Macrophage Interactions. Front Immunol 2021; 12:741502. [PMID: 34671355 PMCID: PMC8521106 DOI: 10.3389/fimmu.2021.741502] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Host innate immune response follows severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and it is the driver of the acute respiratory distress syndrome (ARDS) amongst other inflammatory end-organ morbidities. Such life-threatening coronavirus disease 2019 (COVID-19) is heralded by virus-induced activation of mononuclear phagocytes (MPs; monocytes, macrophages, and dendritic cells). MPs play substantial roles in aberrant immune secretory activities affecting profound systemic inflammation and end-organ malfunctions. All follow the presence of persistent viral components and virions without evidence of viral replication. To elucidate SARS-CoV-2-MP interactions we investigated transcriptomic and proteomic profiles of human monocyte-derived macrophages. While expression of the SARS-CoV-2 receptor, the angiotensin-converting enzyme 2, paralleled monocyte-macrophage differentiation, it failed to affect productive viral infection. In contrast, simple macrophage viral exposure led to robust pro-inflammatory cytokine and chemokine expression but attenuated type I interferon (IFN) activity. Both paralleled dysregulation of innate immune signaling pathways, specifically those linked to IFN. We conclude that the SARS-CoV-2-infected host mounts a robust innate immune response characterized by a pro-inflammatory storm heralding end-organ tissue damage.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Pravin Yeapuri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core, University of Nebraska Medical Center, Omaha, NE, United States
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
32
|
Jafarzadeh A, Jafarzadeh S, Nemati M. Therapeutic potential of ginger against COVID-19: Is there enough evidence? JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [PMCID: PMC8492833 DOI: 10.1016/j.jtcms.2021.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In addition to the respiratory system, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strikes other systems, including the digestive, circulatory, urogenital, and even the central nervous system, as its receptor angiotensin-converting enzyme 2 (ACE2) is expressed in various organs, such as lungs, intestine, heart, esophagus, kidneys, bladder, testis, liver, and brain. Different mechanisms, in particular, massive virus replication, extensive apoptosis and necrosis of the lung-related epithelial and endothelial cells, vascular leakage, hyper-inflammatory responses, overproduction of pro-inflammatory mediators, cytokine storm, oxidative stress, downregulation of ACE2, and impairment of the renin-angiotensin system contribute to the COVID-19 pathogenesis. Currently, COVID-19 is a global pandemic with no specific anti-viral treatment. The favorable capabilities of the ginger were indicated in patients suffering from osteoarthritis, neurodegenerative disorders, rheumatoid arthritis, type 2 diabetes, respiratory distress, liver diseases and primary dysmenorrheal. Ginger or its compounds exhibited strong anti-inflammatory and anti-oxidative influences in numerous animal models. This review provides evidence regarding the potential effects of ginger against SARS-CoV-2 infection and highlights its antiviral, anti-inflammatory, antioxidative, and immunomodulatory impacts in an attempt to consider this plant as an alternative therapeutic agent for COVID-19 treatment.
Collapse
|
33
|
Li D, Sempowski GD, Saunders KO, Acharya P, Haynes BF. SARS-CoV-2 Neutralizing Antibodies for COVID-19 Prevention and Treatment. Annu Rev Med 2021; 73:1-16. [PMID: 34428080 DOI: 10.1146/annurev-med-042420-113838] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Prophylactic and therapeutic drugs are urgently needed to combat coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Over the past year, SARS-CoV-2 neutralizing antibodies have been developed for preventive or therapeutic uses. While neutralizing antibodies target the spike protein, their neutralization potency and breadth vary according to recognition epitopes. Several potent SARS-CoV-2 antibodies have shown degrees of success in preclinical or clinical trials, and the US Food and Drug Administration has issued emergency use authorization for two neutralizing antibody cocktails. Nevertheless, antibody therapy for SARS-CoV-2 still faces potential challenges, including emerging viral variants of concern that have antibody-escape mutations and the potential for antibody-mediated enhancement of infection or inflammation. This review summarizes representative SARS-CoV-2 neutralizing antibodies that have been reported and discusses prospects and challenges for the development of the next generation of COVID-19 preventive or therapeutic antibodies. Expected final online publication date for the Annual Review of Medicine, Volume 73 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA.,Department of Pathology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, North Carolina 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA.,Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
34
|
Boumaza A, Gay L, Mezouar S, Bestion E, Diallo AB, Michel M, Desnues B, Raoult D, La Scola B, Halfon P, Vitte J, Olive D, Mege JL. Monocytes and Macrophages, Targets of Severe Acute Respiratory Syndrome Coronavirus 2: The Clue for Coronavirus Disease 2019 Immunoparalysis. J Infect Dis 2021; 224:395-406. [PMID: 33493287 PMCID: PMC7928817 DOI: 10.1093/infdis/jiab044] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/22/2021] [Indexed: 01/17/2023] Open
Abstract
Background Covid-19 clinical expression is pleiomorphic, severity is related to age and comorbidities such as diabetes and hypertension, and pathophysiology involves aberrant immune activation and lymphopenia. We wondered if the myeloid compartment was affected during Covid-19 and if monocytes and macrophages could be infected by SARS-CoV-2. Methods Monocytes and monocyte-derived macrophages from Covid-19 patients and controls were infected with SARS-CoV-2, and extensively investigated with immunofluorescence, viral RNA extraction and quantification, total RNA extraction followed by reverse transcription and q-PCR using specific primers, supernatant cytokines (IL-10, TNF-α, IL-1β, IFN-β, TGF-β1 and IL-6), flow cytometry. The effect of M1- versus M2-type or no polarization prior to infection was assessed. Results SARS-CoV-2 efficiently infected monocytes and MDMs but their infection is abortive. Infection was associated with immunoregulatory cytokines secretion and the induction of a macrophagic specific transcriptional program characterized by the upregulation of M2-type molecules. In vitro polarization did not account for permissivity to SARS-CoV-2, since M1- and M2-type MDMs were similarly infected. In Covid-19 patients, monocytes exhibited lower counts affecting all subsets, decreased expression of HLA-DR, and increased expression of CD163, irrespective of severity. Conclusion SARS-CoV-2 drives monocytes and macrophages to induce host immunoparalysis for the benefit of Covid-19 progression.
Collapse
Affiliation(s)
- Asma Boumaza
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Laetitia Gay
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France.,ImCheck Therapeutics, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Eloïne Bestion
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France.,Genoscience Pharma, Marseille, France
| | - Aïssatou Bailo Diallo
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Moise Michel
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Benoit Desnues
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Bernard La Scola
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Philippe Halfon
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France.,ImCheck Therapeutics, Marseille, France
| | - Joana Vitte
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France
| | - Daniel Olive
- Centre de recherche en cancérologie de Marseille, Inserm Unité mixte de recherche 1068, Centre National de la Recherche Scientifique Unité mixte de recherche 7258, Institut Paoli Calmettes, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Université, Institut de recherche pour le développement, Assitance publique-hopitaux de Marseille, Microbe, Phylogeny and infection, Marseille, France.,Institut hospitalo-universitaire Méditerranée infection, Marseille, France.,Aix-Marseille Université, Assistance publique-hoptiaux de Marseille, Hopital de la Conception, Laboratoire d'Immunologie, Marseille, France
| |
Collapse
|
35
|
Natarajan H, Xu S, Crowley AR, Butler SE, Weiner JA, Bloch EM, Littlefield K, Benner SE, Shrestha R, Ajayi O, Wieland-alter W, Sullivan D, Shoham S, Quinn TC, Casadevall A, Pekosz A, Redd AD, Tobian AA, Connor RI, Wright PF, Ackerman ME. Antibody Attributes that Predict the Neutralization and Effector Function of Polyclonal Responses to SARS-CoV-2.. [PMID: 34401890 PMCID: PMC8366811 DOI: 10.1101/2021.08.06.21261710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While antibodies provide significant protection from SARS-CoV-2 infection and disease sequelae, the specific attributes of the humoral response that contribute to immunity are incompletely defined. In this study, we employ machine learning to relate characteristics of the polyclonal antibody response raised by natural infection to diverse antibody effector functions and neutralization potency with the goal of generating both accurate predictions of each activity based on antibody response profiles as well as insights into antibody mechanisms of action. To this end, antibody-mediated phagocytosis, cytotoxicity, complement deposition, and neutralization were accurately predicted from biophysical antibody profiles in both discovery and validation cohorts. These predictive models identified SARS-CoV-2-specific IgM as a key predictor of neutralization activity whose mechanistic relevance was supported experimentally by depletion. Validated models of how different aspects of the humoral response relate to antiviral antibody activities suggest desirable attributes to recapitulate by vaccination or other antibody-based interventions.
Collapse
|
36
|
Li D, Edwards RJ, Manne K, Martinez DR, Schäfer A, Alam SM, Wiehe K, Lu X, Parks R, Sutherland LL, Oguin TH, McDanal C, Perez LG, Mansouri K, Gobeil SMC, Janowska K, Stalls V, Kopp M, Cai F, Lee E, Foulger A, Hernandez GE, Sanzone A, Tilahun K, Jiang C, Tse LV, Bock KW, Minai M, Nagata BM, Cronin K, Gee-Lai V, Deyton M, Barr M, Von Holle T, Macintyre AN, Stover E, Feldman J, Hauser BM, Caradonna TM, Scobey TD, Rountree W, Wang Y, Moody MA, Cain DW, DeMarco CT, Denny TN, Woods CW, Petzold EW, Schmidt AG, Teng IT, Zhou T, Kwong PD, Mascola JR, Graham BS, Moore IN, Seder R, Andersen H, Lewis MG, Montefiori DC, Sempowski GD, Baric RS, Acharya P, Haynes BF, Saunders KO. In vitro and in vivo functions of SARS-CoV-2 infection-enhancing and neutralizing antibodies. Cell 2021; 184:4203-4219.e32. [PMID: 34242577 PMCID: PMC8232969 DOI: 10.1016/j.cell.2021.06.021] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
SARS-CoV-2-neutralizing antibodies (NAbs) protect against COVID-19. A concern regarding SARS-CoV-2 antibodies is whether they mediate disease enhancement. Here, we isolated NAbs against the receptor-binding domain (RBD) or the N-terminal domain (NTD) of SARS-CoV-2 spike from individuals with acute or convalescent SARS-CoV-2 or a history of SARS-CoV infection. Cryo-electron microscopy of RBD and NTD antibodies demonstrated function-specific modes of binding. Select RBD NAbs also demonstrated Fc receptor-γ (FcγR)-mediated enhancement of virus infection in vitro, while five non-neutralizing NTD antibodies mediated FcγR-independent in vitro infection enhancement. However, both types of infection-enhancing antibodies protected from SARS-CoV-2 replication in monkeys and mice. Three of 46 monkeys infused with enhancing antibodies had higher lung inflammation scores compared to controls. One monkey had alveolar edema and elevated bronchoalveolar lavage inflammatory cytokines. Thus, while in vitro antibody-enhanced infection does not necessarily herald enhanced infection in vivo, increased lung inflammation can rarely occur in SARS-CoV-2 antibody-infused macaques.
Collapse
Affiliation(s)
- Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kartik Manne
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Laura L Sutherland
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas H Oguin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Lautaro G Perez
- Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sophie M C Gobeil
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Katarzyna Janowska
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Megan Kopp
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Fangping Cai
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Esther Lee
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Andrew Foulger
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Giovanna E Hernandez
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Aja Sanzone
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kedamawit Tilahun
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chuancang Jiang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Longping V Tse
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenneth Cronin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Victoria Gee-Lai
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Margaret Deyton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tarra Von Holle
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Andrew N Macintyre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Erica Stover
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Blake M Hauser
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Trevor D Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - C Todd DeMarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Christopher W Woods
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Elizabeth W Petzold
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | | | | | | | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
37
|
Leander RN, Wu Y, Ding W, Nelson DE, Sinkala Z. A model of the innate immune response to SARS-CoV-2 in the alveolar epithelium. ROYAL SOCIETY OPEN SCIENCE 2021; 8:210090. [PMID: 34430043 PMCID: PMC8355678 DOI: 10.1098/rsos.210090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/19/2021] [Indexed: 05/15/2023]
Abstract
We present a differential equation model of the innate immune response to SARS-CoV-2 within the alveolar epithelium. Critical determinants of the viral dynamics and host response, including type I and type II alveolar epithelial cells, interferons, chemokines, toxins and innate immune cells, are included. We estimate model parameters, compute the within-host basic reproductive number, and study the impacts of therapies, prophylactics, and host/pathogen variability on the course of the infection. Model simulations indicate that the innate immune response suppresses the infection and enables the alveolar epithelium to partially recover. While very robust antiviral therapy controls the infection and enables the epithelium to heal, moderate therapy is of limited benefit. Meanwhile interferon therapy is predicted to reduce viral load but exacerbate tissue damage. The deleterious effects of interferon therapy are especially apparent late in the infection. Individual variation in ACE2 expression, epithelial cell interferon production, and SARS-CoV-2 spike protein binding affinity are predicted to significantly impact prognosis.
Collapse
Affiliation(s)
- R. N. Leander
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro 37132-0002, USA
| | - Y. Wu
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro 37132-0002, USA
| | - W. Ding
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro 37132-0002, USA
| | - D. E. Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro 37132-0002, USA
| | - Z. Sinkala
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro 37132-0002, USA
| |
Collapse
|
38
|
Huang C. Pathogenesis of Coronaviruses Through Human Monocytes and Tissue Macrophages. Viral Immunol 2021; 34:597-606. [PMID: 34297627 DOI: 10.1089/vim.2021.0038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Coronaviruses (CoVs) contribute significantly to the burden of respiratory diseases, frequently as upper respiratory tract infections. Recent emergence of novel coronaviruses in the last few decades has highlighted the potential transmission, disease, and mortality related to these viruses. In this literature review, we shall explore the disease-causing mechanism of the virus through human monocytes and macrophages. Common strains will be discussed; however, this review will center around coronaviruses responsible for epidemics, namely severe acute respiratory syndrome coronavirus (SARS-CoV)-1 and -2 and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). Macrophages are key players in the immune system and have been found to play a role in the pathogenesis of lethal coronaviruses. In physiology, they are white blood cells that engulf and digest cellular debris, foreign substances, and microbes. They play a critical role in innate immunity and help initiate adaptive immunity. Human coronaviruses utilize various mechanisms to undermine the innate immune response through its interaction with macrophages and monocytes. It is capable of entering immune cells through DPP4 (dipeptidyl-peptidase 4) receptors and antibody-dependent enhancement, delaying initial interferon response which supports robust viral replication. Pathogenesis includes triggering the production of overwhelming pro-inflammatory cytokines that attract other immune cells to the site of infection, which propagate prolonged pro-inflammatory response. The virus has also been found to suppress the release of anti-inflammatory mediators such as IL-10, leading to an aberrant inflammatory response. Elevated serum cytokines are also believed to contribute to pathological features seen in severe disease such as coagulopathy, acute lung injury, and multiorgan failure.
Collapse
Affiliation(s)
- Chenghao Huang
- Medical School, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
39
|
Knoll R, Schultze JL, Schulte-Schrepping J. Monocytes and Macrophages in COVID-19. Front Immunol 2021; 12:720109. [PMID: 34367190 PMCID: PMC8335157 DOI: 10.3389/fimmu.2021.720109] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a contagious viral disease caused by SARS-CoV-2 that led to an ongoing pandemic with massive global health and socioeconomic consequences. The disease is characterized primarily, but not exclusively, by respiratory clinical manifestations ranging from mild common cold symptoms, including cough and fever, to severe respiratory distress and multi-organ failure. Macrophages, a heterogeneous group of yolk-sac derived, tissue-resident mononuclear phagocytes of complex ontogeny present in all mammalian organs, play critical roles in developmental, homeostatic and host defense processes with tissue-dependent plasticity. In case of infection, they are responsible for early pathogen recognition, initiation and resolution of inflammation, as well as repair of tissue damage. Monocytes, bone-marrow derived blood-resident phagocytes, are recruited under pathological conditions such as viral infections to the affected tissue to defend the organism against invading pathogens and to aid in efficient resolution of inflammation. Given their pivotal function in host defense and the potential danger posed by their dysregulated hyperinflammation, understanding monocyte and macrophage phenotypes in COVID-19 is key for tackling the disease's pathological mechanisms. Here, we outline current knowledge on monocytes and macrophages in homeostasis and viral infections and summarize concepts and key findings on their role in COVID-19. While monocytes in the blood of patients with moderate COVID-19 present with an inflammatory, interferon-stimulated gene (ISG)-driven phenotype, cellular dysfunction epitomized by loss of HLA-DR expression and induction of S100 alarmin expression is their dominant feature in severe disease. Pulmonary macrophages in COVID-19 derived from infiltrating inflammatory monocytes are in a hyperactivated state resulting in a detrimental loop of pro-inflammatory cytokine release and recruitment of cytotoxic effector cells thereby exacerbating tissue damage at the site of infection.
Collapse
Affiliation(s)
- Rainer Knoll
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L. Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Jonas Schulte-Schrepping
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
40
|
Abdelmoaty M, Yeapuri P, Machhi J, Olson K, Shahjin F, Zhou Y, Jingjing L, Pandey K, Acharya A, Byrareddy S, Mosley L, Gendelman H. Defining the Immune Responses for SARS-CoV-2-Human Macrophage Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.07.07.449660. [PMID: 34268510 PMCID: PMC8282098 DOI: 10.1101/2021.07.07.449660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Host innate immune response follows severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and it is the driver of the acute respiratory distress syndrome (ARDS) amongst other inflammatory end-organ morbidities. Such life-threatening coronavirus disease 2019 (COVID-19) is heralded by virus-induced activation of mononuclear phagocytes (MPs; monocytes, macrophages, and dendritic cells). MPs play substantial roles in aberrant immune secretory activities affecting profound systemic inflammation and end organ malfunctions. All follow an abortive viral infection. To elucidate SARS-CoV-2-MP interactions we investigated transcriptomic and proteomic profiles of human monocyte-derived macrophages. While expression of the SARS-CoV-2 receptor, the angiotensin-converting enzyme 2, paralleled monocyte-macrophage differentiation it failed to affect productive viral infection. In contrast, simple macrophage viral exposure led to robust pro-inflammatory cytokine and chemokine expression but attenuated type I interferon (IFN) activity. Both paralleled dysregulation of innate immune signaling pathways specifically those linked to IFN. We conclude that the SARS-CoV-2-infected host mounts a robust innate immune response characterized by a pro-inflammatory storm heralding consequent end-organ tissue damage.
Collapse
|
41
|
Abstract
BACKGROUND SARS-CoV-2, a coronavirus (CoV), is known to cause acute respiratory distress syndrome, and a number of non-respiratory complications, particularly in older male patients with prior health conditions, such as obesity, diabetes and hypertension. These prior health conditions are associated with vascular dysfunction, and the CoV disease 2019 (COVID-19) complications include multiorgan failure and neurological problems. While the main route of entry into the body is inhalation, this virus has been found in many tissues, including the choroid plexus and meningeal vessels, and in neurons and CSF. MAIN BODY We reviewed SARS-CoV-2/COVID-19, ACE2 distribution and beneficial effects, the CNS vascular barriers, possible mechanisms by which the virus enters the brain, outlined prior health conditions (obesity, hypertension and diabetes), neurological COVID-19 manifestation and the aging cerebrovascualture. The overall aim is to provide the general reader with a breadth of information on this type of virus and the wide distribution of its main receptor so as to better understand the significance of neurological complications, uniqueness of the brain, and the pre-existing medical conditions that affect brain. The main issue is that there is no sound evidence for large flux of SARS-CoV-2 into brain, at present, compared to its invasion of the inhalation pathways. CONCLUSIONS While SARS-CoV-2 is detected in brains from severely infected patients, it is unclear on how it gets there. There is no sound evidence of SARS-CoV-2 flux into brain to significantly contribute to the overall outcomes once the respiratory system is invaded by the virus. The consensus, based on the normal route of infection and presence of SARS-CoV-2 in severely infected patients, is that the olfactory mucosa is a possible route into brain. Studies are needed to demonstrate flux of SARS-CoV-2 into brain, and its replication in the parenchyma to demonstrate neuroinvasion. It is possible that the neurological manifestations of COVID-19 are a consequence of mainly cardio-respiratory distress and multiorgan failure. Understanding potential SARS-CoV-2 neuroinvasion pathways could help to better define the non-respiratory neurological manifestation of COVID-19.
Collapse
Affiliation(s)
- Conor McQuaid
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| | - Molly Brady
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| | - Rashid Deane
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| |
Collapse
|
42
|
Chazal N. Coronavirus, the King Who Wanted More Than a Crown: From Common to the Highly Pathogenic SARS-CoV-2, Is the Key in the Accessory Genes? Front Microbiol 2021; 12:682603. [PMID: 34335504 PMCID: PMC8317507 DOI: 10.3389/fmicb.2021.682603] [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: 03/18/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that emerged in late 2019, is the etiologic agent of the current "coronavirus disease 2019" (COVID-19) pandemic, which has serious health implications and a significant global economic impact. Of the seven human coronaviruses, all of which have a zoonotic origin, the pandemic SARS-CoV-2, is the third emerging coronavirus, in the 21st century, highly pathogenic to the human population. Previous human coronavirus outbreaks (SARS-CoV-1 and MERS-CoV) have already provided several valuable information on some of the common molecular and cellular mechanisms of coronavirus infections as well as their origin. However, to meet the new challenge caused by the SARS-CoV-2, a detailed understanding of the biological specificities, as well as knowledge of the origin are crucial to provide information on viral pathogenicity, transmission and epidemiology, and to enable strategies for therapeutic interventions and drug discovery. Therefore, in this review, we summarize the current advances in SARS-CoV-2 knowledges, in light of pre-existing information of other recently emerging coronaviruses. We depict the specificity of the immune response of wild bats and discuss current knowledge of the genetic diversity of bat-hosted coronaviruses that promotes viral genome expansion (accessory gene acquisition). In addition, we describe the basic virology of coronaviruses with a special focus SARS-CoV-2. Finally, we highlight, in detail, the current knowledge of genes and accessory proteins which we postulate to be the major keys to promote virus adaptation to specific hosts (bat and human), to contribute to the suppression of immune responses, as well as to pathogenicity.
Collapse
Affiliation(s)
- Nathalie Chazal
- Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
43
|
Idris A, Davis A, Supramaniam A, Acharya D, Kelly G, Tayyar Y, West N, Zhang P, McMillan CLD, Soemardy C, Ray R, O'Meally D, Scott TA, McMillan NAJ, Morris KV. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. Mol Ther 2021; 29:2219-2226. [PMID: 33992805 PMCID: PMC8118699 DOI: 10.1016/j.ymthe.2021.05.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 01/16/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective small interfering RNA (siRNA) therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle (LNP) delivery system. Multiple siRNAs targeting highly conserved regions of the SARS-CoV-2 virus were screened, and three candidate siRNAs emerged that effectively inhibit the virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel LNP formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.
Collapse
Affiliation(s)
- Adi Idris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Alicia Davis
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA; Irell & Manella Graduate School of Biological Sciences at the City of Hope, Duarte, CA 91010, USA
| | - Aroon Supramaniam
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Dhruba Acharya
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Gabrielle Kelly
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Nic West
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Ping Zhang
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Citradewi Soemardy
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Roslyn Ray
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Denis O'Meally
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Tristan A Scott
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Nigel A J McMillan
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia.
| | - Kevin V Morris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia; Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
44
|
Ropa J, Trinh T, Aljoufi A, Broxmeyer HE. Consequences of coronavirus infections for primitive and mature hematopoietic cells: new insights and why it matters. Curr Opin Hematol 2021; 28:231-242. [PMID: 33656463 PMCID: PMC8269959 DOI: 10.1097/moh.0000000000000645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW In recent history there have been three outbreaks of betacoronavirus infections in humans, with the most recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2; causing Coronavirus disease 2019 [COVID-19]) outbreak leading to over two million deaths, with a rapidly rising death toll. Much remains unknown about host cells and tissues affected by coronavirus infections, including the hematopoietic system. Here, we discuss the recent findings examining effects that coronavirus infection or exposure has on hematopoietic cells and the clinical implications for these effects. RECENT FINDINGS Recent studies have centered on SARS-CoV-2, demonstrating that hematopoietic stem and progenitor cells and mature immune cells may be susceptible to infection and are impacted functionally by exposure to SARS-CoV-2 Spike protein. These findings have important implications regarding hematologic complications arising from COVID-19 and other coronavirus-induced disease, which we discuss here. SUMMARY Infection with coronaviruses sometimes leads to hematologic complications in patients, and these hematologic complications are associated with poorer prognosis. These hematologic complications may be caused by coronavirus direct infection or impact on primitive hematopoietic cells or mature immune cells, by indirect effects on these cells, or by a combination thereof. It is important to understand how hematologic complications arise in order to seek new treatments to improve patient outcomes.
Collapse
Affiliation(s)
- James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Thao Trinh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Arafat Aljoufi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hal E. Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
45
|
Geng T, Keilich S, Tafas T, Wang P. Early detection of SARS-CoV-2 in circulating immune cells in a mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.06.30.450531. [PMID: 34230928 PMCID: PMC8259904 DOI: 10.1101/2021.06.30.450531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
SARS-CoV-2 infects the respiratory tract, lung and then other organs. However, its pathogenesis remains largely unknown. We used RareScope™ Fluorescence Light Sheet Microscopy (FLSM) and fluorescent in situ hybridization of RNA (RNA-FISH) to detect SARS-CoV-2 RNA and dissemination kinetics in mouse blood circulation. By RNA-FISH, we found that SARS-CoV-2 RNA-positive leukocytes, including CD11c cells, appeared as early as one day after infection and continued through day 10 post infection. Our data suggest that SARS-CoV-2-permissive leukocytes contribute to systemic viral dissemination, and RNA-FISH combined with FLSM can be utilized as a sensitive tool for SARS-CoV-2 detection in blood specimens.
Collapse
Affiliation(s)
- Tingting Geng
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
46
|
Study of the Plasma and Buffy Coat in Patients with SARS-CoV-2 Infection-A Preliminary Report. Pathogens 2021; 10:pathogens10070805. [PMID: 34201967 PMCID: PMC8308615 DOI: 10.3390/pathogens10070805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022] Open
Abstract
The pandemic caused by the SARS-CoV-2 infection affects many aspects of public health knowledge, science, and practice around the world. Several studies have shown that SARS-CoV-2 RNA in plasma seems to be associated with a worse prognosis of COVID-19. In the present study, we investigated plasma and buffy RNA in patients with COVID-19 to determine its prognostic value. A prospective study was carried out in patients hospitalized for COVID-19, in which RNA was analyzed in plasma and the buffy coat. Morphological and immunohistochemical studies were used to detect the presence of SARS-CoV-2 in the buffy coat. In COVID-19 patients, the obtained RNA concentration in plasma was 448.3 ± 31.30 ng/mL. Of all the patients with positive plasma tests for SARS-CoV-2, 46.15% died from COVID-19. In four cases, tests revealed that SARS-CoV-2 was present in the buffy coat. Abnormal morphology of monocytes, lymphocytes and neutrophils was found. An immunohistochemical study showed positivity in mononuclear cells and platelets. Our results suggest that SARS-CoV-2 is present in the plasma. This facilitates viral dissemination and migration to specific organs, where SARS-CoV-2 infects target cells by binding to their receptors. In our study, the presence of plasma SARS-CoV-2 RNA was correlated with worse prognoses.
Collapse
|
47
|
Piri SM, Edalatfar M, Shool S, Jalalian MN, Tavakolpour S. A systematic review on the recurrence of SARS-CoV-2 virus: frequency, risk factors, and possible explanations. Infect Dis (Lond) 2021; 53:315-324. [PMID: 33508989 PMCID: PMC7852280 DOI: 10.1080/23744235.2020.1871066] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Since late 2019, SARS-CoV-2 which leads to coronavirus disease 2019 (COVID-19), has caused thousands of deaths. There are some pieces of evidence that SARS-CoV-2 genome could be re-detectable in recovered patients. METHODS We performed a systematic review in the PubMed/Medline database to address the risk of SARS-CoV-2 recurrence. The last update was for 20 November 2020. Among the 1178 initially found articles, 66 met the inclusion criteria and were considered. FINDINGS In total, 1128 patients with at least one-time recurrence of SARS-CoV-2 were included. Recurrence rate has been reported between 2.3% and 21.4% in cohort studies, within a mean of 20 (ranged 1-98) days after discharge; younger patients are being affected more. Following the second course of disease, the disease severity decreased or remained unchanged in 97.3% while it increased in 2.6%. Anti-SARS-CoV-2 IgG and IgM were positive in 11-95% and 58.8-100%, respectively. Based on the literature, three possibilities include reactivation of previous disease, reinfection with the same virus, and false negative, which have been discussed in details. CONCLUSION There is a relatively notable risk of disease recurrence in previously recovered patients, even those who are immunised against the virus. More studies are required to clarify the underlying cause of this phenomenon.
Collapse
Affiliation(s)
- Seyed Mohammad Piri
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Edalatfar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Shool
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheil Tavakolpour
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA,CONTACT Soheil Tavakolpour Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Degauque N, Haziot A, Brouard S, Mooney N. Endothelial cell, myeloid, and adaptive immune responses in SARS-CoV-2 infection. FASEB J 2021; 35:e21577. [PMID: 33831263 PMCID: PMC8250117 DOI: 10.1096/fj.202100024r] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is an emerging respiratory pathogen that has rapidly spread in human populations. Severe forms of infection associate cytokine release syndrome and acute lung injury due to hyperinflammatory responses even though virus clearance is achieved. Key components of inflammation include immune cell recruitment in infected tissues, a step which is under the control of endothelial cells. Here, we review endothelial cell responses in inflammation and infection due to SARS-CoV-2 together with phenotypic and functional alterations of monocytes, T and B lymphocytes with which they interact. We surmise that endothelial cells function as an integrative and active platform for the various cells recruited, where fine tuning of immune responses takes place and which provides opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Nicolas Degauque
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Alain Haziot
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| | - Sophie Brouard
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Nuala Mooney
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| |
Collapse
|
49
|
Natarajan H, Crowley AR, Butler SE, Xu S, Weiner JA, Bloch EM, Littlefield K, Wieland-Alter W, Connor RI, Wright PF, Benner SE, Bonny TS, Laeyendecker O, Sullivan D, Shoham S, Quinn TC, Larman HB, Casadevall A, Pekosz A, Redd AD, Tobian AAR, Ackerman ME. Markers of Polyfunctional SARS-CoV-2 Antibodies in Convalescent Plasma. mBio 2021; 12:e00765-21. [PMID: 33879585 PMCID: PMC8092262 DOI: 10.1128/mbio.00765-21] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
Convalescent plasma is a promising therapy for coronavirus disease 2019 (COVID-19), but the antibody characteristics that contribute to efficacy remain poorly understood. This study analyzed plasma samples from 126 eligible convalescent blood donors in addition to 15 naive individuals, as well as an additional 20 convalescent individuals as a validation cohort. Multiplexed Fc Array binding assays and functional antibody response assays were utilized to evaluate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody composition and activity. Donor convalescent plasma samples contained a range of antibody cell- and complement-mediated effector functions, indicating the diverse antiviral activity of humoral responses observed among recovered individuals. In addition to viral neutralization, convalescent plasma samples contained antibodies capable of mediating such Fc-dependent functions as complement activation, phagocytosis, and antibody-dependent cellular cytotoxicity against SARS-CoV-2. Plasma samples from a fraction of eligible donors exhibited high activity across all activities evaluated. These polyfunctional plasma samples could be identified with high accuracy with even single Fc Array features, whose correlation with polyfunctional activity was confirmed in the validation cohort. Collectively, these results expand understanding of the diversity of antibody-mediated antiviral functions associated with convalescent plasma, and the polyfunctional antiviral functions suggest that it could retain activity even when its neutralizing capacity is reduced by mutations in variant SARS-CoV-2.IMPORTANCE Convalescent plasma has been deployed globally as a treatment for COVID-19, but efficacy has been mixed. Better understanding of the antibody characteristics that may contribute to its antiviral effects is important for this intervention as well as offer insights into correlates of vaccine-mediated protection. Here, a survey of convalescent plasma activities, including antibody neutralization and diverse effector functions, was used to define plasma samples with broad activity profiles. These polyfunctional plasma samples could be reliably identified in multiple cohorts by multiplex assay, presenting a widely deployable screening test for plasma selection and investigation of vaccine-elicited responses.
Collapse
Affiliation(s)
- Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
| | - Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
| | - Savannah E Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
| | - Shiwei Xu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Wendy Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Ruth I Connor
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Sarah E Benner
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Tania S Bonny
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Thomas C Quinn
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - H Benjamin Larman
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
50
|
Idris A, Davis A, Supramaniam A, Acharya D, Kelly G, Tayyar Y, West N, Zhang P, McMillan CLD, Soemardy C, Ray R, O'Meally D, Scott TA, McMillan NAJ, Morris KV. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33907744 DOI: 10.1101/2021.04.19.440531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective siRNA therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle delivery system. Multiple small-interfering RNAs (siRNAs) targeting highly conserved regions of the SARS-CoV-2 virus were screened and three candidate siRNAs emerged that effectively inhibit virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel lipid nanoparticle formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.
Collapse
|