1
|
Niayesh-Mehr R, Kalantar M, Bontempi G, Montaldo C, Ebrahimi S, Allameh A, Babaei G, Seif F, Strippoli R. The role of epithelial-mesenchymal transition in pulmonary fibrosis: lessons from idiopathic pulmonary fibrosis and COVID-19. Cell Commun Signal 2024; 22:542. [PMID: 39538298 PMCID: PMC11558984 DOI: 10.1186/s12964-024-01925-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the tremendous advancements in the knowledge of the pathophysiology and clinical aspects of SARS-CoV-2 infection, still many issues remain unanswered, especially in the long-term effects. Mounting evidence suggests that pulmonary fibrosis (PF) is one of the most severe complications associated with COVID-19. Therefore, understanding the molecular mechanisms behind its development is helpful to develop successful therapeutic strategies. Epithelial to mesenchymal transition (EMT) and its cell specific variants endothelial to mesenchymal transition (EndMT) and mesothelial to mesenchymal transition (MMT) are physio-pathologic cellular reprogramming processes induced by several infectious, inflammatory and biomechanical stimuli. Cells undergoing EMT acquire invasive, profibrogenic and proinflammatory activities by secreting several extracellular mediators. Their activity has been implicated in the pathogenesis of PF in a variety of lung disorders, including idiopathic pulmonary fibrosis (IPF) and COVID-19. Aim of this article is to provide an updated survey of the cellular and molecular mechanisms, with emphasis on EMT-related processes, implicated in the genesis of PF in IFP and COVID-19.
Collapse
Affiliation(s)
- Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Kalantar
- Department of Occupational Health, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Saeedeh Ebrahimi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Faezeh Seif
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
2
|
Zhang WY, Wen L, Du L, Liu TT, Sun Y, Chen YZ, Lu YX, Cheng XC, Sun HY, Xiao FJ, Wang LS. S-RBD-modified and miR-486-5p-engineered exosomes derived from mesenchymal stem cells suppress ferroptosis and alleviate radiation-induced lung injury and long-term pulmonary fibrosis. J Nanobiotechnology 2024; 22:662. [PMID: 39462403 PMCID: PMC11515248 DOI: 10.1186/s12951-024-02830-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Radiation-induced lung injury (RILI) is associated with alveolar epithelial cell death and secondary fibrosis in injured lung. Mesenchymal stem cell (MSC)-derived exosomes have regenerative effect against lung injury and the potential to intervene of RILI. However, their intervention efficacy is limited because they lack lung targeting characters and do not carry sufficient specific effectors. SARS-CoV-2 spike glycoprotein (SARS-CoV-2-S-RBD) binds angiotensin-converting enzyme 2 (ACE2) receptor and mediates interaction with host cells. MiR-486-5p is a multifunctional miRNA with angiogenic and antifibrotic potential and acts as an effector in MSC-derived exosomes. Ferroptosis is a form of cell death associated with radiation injury, its roles and mechanisms in RILI remain unclear. In this study, we developed an engineered MSC-derived exosomes with SARS-CoV-2-S-RBD- and miR-486-5p- modification and investigated their intervention effects on RIPF and action mechanisms via suppression of epithelial cell ferroptosis. RESULTS Adenovirus-mediated gene modification led to miR-486-5p overexpression in human umbilical cord MSC exosomes (p < 0.05), thereby constructing miR-486-5p engineered MSC exosomes (miR-486-MSC-Exo). MiR-486-MSC-Exo promoted the proliferation and migration of irradiated mouse lung epithelial (MLE-12) cells in vitro and inhibited RILI in vivo (all p < 0.05). MiR-486-MSC-Exo suppressed ferroptosis in MLE-12 cells, and an in vitro assay revealed that the expression of fibrosis-related genes is up-regulated following ferroptosis (both p < 0.05). MiR-486-MSC-Exo reversed the up-regulated expression of fibrosis-related genes induced by TGF-β1 in vitro and improved pathological fibrosis in RIPF mice in vivo (all p < 0.05). SARS-CoV-2-S-RBD-modified and miR-486-5p-engineered MSC exosomes (miR-486-RBD-MSC-Exo) were also constructed, and the distribution of DiR dye-labeled miR-486-RBD-MSC-Exo in hACE2CKI/CKI Sftpc-Cre+ mice demonstrated long-term retention in the lung (p < 0.05). MiR-486-RBD-MSC-Exo significantly improved the survival rate and pathological changes in hACE2CKI/CKI Sftpc-Cre+ RIPF mice (all p < 0.05). Furthermore, miR-486-MSC-Exo exerted anti-fibrotic effects via targeted SMAD2 inhibition and Akt phosphorylation activation (p < 0.05). CONCLUSIONS Engineered MSC exosomes with SARS-CoV-2-S-RBD- and miR-486-5p-modification were developed. MiR-486-RBD-MSC-Exo suppressed ferroptosis and fibrosis of MLE-12 cells in vitro, and alleviated RILI and long-term RIPF in ACE2 humanized mice in vivo. MiR-486-MSC-Exo exerted anti-fibrotic effects via SMAD2 inhibition and Akt activation. This study provides a potential approach for RIPF intervention.
Collapse
Affiliation(s)
- Wei-Yuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Li Wen
- School of Nursing, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Li Du
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Ting Ting Liu
- Department of Pulmonary and Critical Care Medicine, The Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Sun
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Yi-Zhu Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Yu-Xin Lu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Xiao-Chen Cheng
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Hui-Yan Sun
- Yanda Medical Research Institute, Hebei Yanda Hospital, Langfang, 065201, China
| | - Feng-Jun Xiao
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Li-Sheng Wang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- School of Nursing, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
3
|
Vontetsianos A, Chynkiamis N, Anagnostopoulou C, Lekka C, Zaneli S, Anagnostopoulos N, Rovina N, Kampolis CF, Papaioannou AI, Kaltsakas G, Vogiatzis I, Stratakos G, Bakakos P, Koulouris N. Small Airways Dysfunction and Lung Hyperinflation in Long COVID-19 Patients as Potential Mechanisms of Persistent Dyspnoea. Adv Respir Med 2024; 92:329-337. [PMID: 39311110 PMCID: PMC11417746 DOI: 10.3390/arm92050031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Reticulation, ground glass opacities and post-infection bronchiectasis are present three months following hospitalisation in patients recovering from SARS-CoV-2 infection and are associated with the severity of acute infection. However, scarce data exist on small airways impairment and lung hyperinflation in patients with long COVID-19. AIM To evaluate small airways function and lung hyperinflation in previously hospitalised patients with long COVID-19 and their association with post-COVID-19 breathlessness. METHODS In total, 33 patients (mean ± SD, 53 ± 11 years) with long COVID-19 were recruited 149 ± 90 days following hospital discharge. Pulmonary function tests were performed and lung hyperinflation was defined as RV/TLC ≥ 40%. Small airways function was evaluated by measuring the closing volume (CV) and closing capacity (CC) using the single-breath nitrogen washout technique (SBN2W). RESULTS CC was 115 ± 28% pred. and open capacity (OC) was 90 ± 19. CC was abnormal in 13 patients (39%), CV in 2 patients (6.1%) and OC in 9 patients (27%). Lung hyperinflation was present in 15 patients, whilst the mean mMRC score was 2.2 ± 1.0. Lung hyperinflation was associated with CC (r = 0.772, p = 0.001), OC (r = 0.895, p = 0.001) and mMRC (r = 0.444, p = 0.010). CONCLUSIONS Long COVID-19 patients present with small airways dysfunction and lung hyperinflation, which is associated with persistent dyspnoea, following hospitalisation.
Collapse
Affiliation(s)
- Angelos Vontetsianos
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Nikolaos Chynkiamis
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
- Thorax Research Foundation, 11521 Athens, Greece
| | - Christina Anagnostopoulou
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Christiana Lekka
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Stavrina Zaneli
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Nektarios Anagnostopoulos
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Nikoleta Rovina
- 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christos F. Kampolis
- Department of Emergency Medicine, “Hippokration” General Hospital of Athens, 11527 Athens, Greece;
| | - Andriana I. Papaioannou
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Georgios Kaltsakas
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
- Lane Fox Respiratory Service, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK
- Centre of Human and Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK
| | - Ioannis Vogiatzis
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
- Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University Newcastle, Newcastle upon Tyne NE1 8ST, UK
| | - Grigorios Stratakos
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Petros Bakakos
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| | - Nikolaos Koulouris
- Rehabilitation Unit, 1st Respiratory Medicine Department, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.V.); (C.A.); (C.L.); (S.Z.); (N.A.); (A.I.P.); (G.K.); (I.V.); (G.S.); (P.B.); (N.K.)
| |
Collapse
|
4
|
Fernandez SA, Pelaez-Prestel HF, Ras-Carmona A, Mozas-Gutierrez J, Reyes-Manzanas R, Reche PA. Eucalyptus Essential Oil Inhibits Cell Infection by SARS-CoV-2 Spike Pseudotyped Lentivirus. Biomedicines 2024; 12:1885. [PMID: 39200349 PMCID: PMC11351113 DOI: 10.3390/biomedicines12081885] [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: 07/19/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a public health concern due to infections with new SARS-CoV-2 variants. Therefore, finding effective preventive and therapeutic treatments against all SARS-CoV-2 variants is of great interest. In this study, we examined the capacity of eucalyptus essential oil (EEO) and eucalyptol (EOL) to prevent SARS-CoV-2 infection, using as a model SARS-CoV-2 Spike pseudotyped lentivirus (SARS-CoV-2 pseudovirus) and 293T cells transfected with human angiotensin-converting enzyme 2 (hACE2-293T cells). First, we determined the cytotoxicity of EEO and EOL using the MTT colorimetric assay, selecting non-cytotoxic concentrations ≤ 0.1% (v/v) for further analysis. Subsequently, we evaluated the capacity of EEO and EOL in cell cultures to preclude infection of hACE2-293T cells by SARS-CoV-2 pseudovirus, using a luciferase-based assay. We found that EEO and EOL significantly reduced SARS-CoV-2 pseudovirus infection, obtaining IC50 values of 0.00895% and 0.0042% (v/v), respectively. Likewise, EEO and EOL also reduced infection by vesicular stomatitis virus (VSV) pseudovirus, although higher concentrations were required. Hence, EEO and EOL may be able to inhibit SARS-CoV-2 infection, at least partially, through a Spike-independent pathway, supporting the implementation of aromatherapy with these agents as a cost-effective antiviral measure.
Collapse
Affiliation(s)
| | | | | | | | - Raquel Reyes-Manzanas
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, S/N, 28040 Madrid, Spain; (S.A.F.); (H.F.P.-P.); (A.R.-C.); (J.M.-G.)
| | - Pedro A. Reche
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, S/N, 28040 Madrid, Spain; (S.A.F.); (H.F.P.-P.); (A.R.-C.); (J.M.-G.)
| |
Collapse
|
5
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
6
|
Magnen M, You R, Rao AA, Davis RT, Rodriguez L, Bernard O, Simoneau CR, Hysenaj L, Hu KH, Maishan M, Conrad C, Gbenedio OM, Samad B, Consortium TUCSFCOMET, Love C, Woodruff PG, Erle DJ, Hendrickson CM, Calfee CS, Matthay MA, Roose JP, Sil A, Ott M, Langelier CR, Krummel MF, Looney MR. Immediate myeloid depot for SARS-CoV-2 in the human lung. SCIENCE ADVANCES 2024; 10:eadm8836. [PMID: 39083602 PMCID: PMC11290487 DOI: 10.1126/sciadv.adm8836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/20/2024] [Indexed: 08/02/2024]
Abstract
In the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, epithelial populations in the distal lung expressing Angiotensin-converting enzyme 2 (ACE2) are infrequent, and therefore, the model of viral expansion and immune cell engagement remains incompletely understood. Using human lungs to investigate early host-viral pathogenesis, we found that SARS-CoV-2 had a rapid and specific tropism for myeloid populations. Human alveolar macrophages (AMs) reliably expressed ACE2 allowing both spike-ACE2-dependent viral entry and infection. In contrast to Influenza A virus, SARS-CoV-2 infection of AMs was productive, amplifying viral titers. While AMs generated new viruses, the interferon responses to SARS-CoV-2 were muted, hiding the viral dissemination from specific antiviral immune responses. The reliable and veiled viral depot in myeloid cells in the very early phases of SARS-CoV-2 infection of human lungs enables viral expansion in the distal lung and potentially licenses subsequent immune pathologies.
Collapse
Affiliation(s)
- Mélia Magnen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ran You
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun A. Rao
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ryan T. Davis
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lauren Rodriguez
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Olivier Bernard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Camille R. Simoneau
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lisiena Hysenaj
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenneth H. Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mazharul Maishan
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Catharina Conrad
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Oghenekevwe M. Gbenedio
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - The UCSF COMET Consortium
- All UCSF COMET Consortium collaborators are affiliated with the University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christina Love
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Prescott G. Woodruff
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David J. Erle
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carolyn M. Hendrickson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carolyn S. Calfee
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael A. Matthay
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Melanie Ott
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Charles R. Langelier
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
7
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
8
|
Tsuji M, Kondo M, Sato Y, Miyoshi A, Kobayashi F, Arimura K, Yamashita K, Morimoto S, Yanagisawa N, Ichihara A, Tagaya E. Serum VEGF-A levels on admission in COVID-19 patients correlate with SP-D and neutrophils, reflecting disease severity: A prospective study. Cytokine 2024; 178:156583. [PMID: 38554499 DOI: 10.1016/j.cyto.2024.156583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND AND OBJECTIVE The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant global morbidity and mortality. This study aimed to investigate the clinical significance of serum vascular endothelial growth factor A (VEGF-A) in COVID-19 patients and its association with disease severity and pulmonary injury. METHODS We prospectively collected data from 71 hospitalized COVID-19 patients between June 2020 and January 2021. Patients were classified as either mild or severe based on their oxygen requirements during hospitalization. Serum VEGF-A levels were measured using an ELISA kit. RESULTS In comparison to mild cases, significantly elevated serum VEGF-A levels were observed in severe COVID-19 patients. Furthermore, VEGF-A levels exhibited a positive correlation with white blood cell count, neutrophil count, and lymphocyte count. Notably, serum surfactant protein-D (SP-D), an indicator of alveolar epithelial cell damage, was significantly higher in patients with elevated VEGF-A levels. CONCLUSION These results suggest that elevated serum VEGF-A levels could serve as a prognostic biomarker for COVID-19 as it is indicative of alveolar epithelial cell injury caused by SARS-CoV-2 infection. Additionally, we observed a correlation between VEGF-A and neutrophil activation, which plays a role in the immune response during endothelial cell injury, indicating a potential involvement of angiogenesis in disease progression. Further research is needed to elucidate the underlying mechanisms of VEGF-A elevation in COVID-19.
Collapse
Affiliation(s)
- Mayoko Tsuji
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| | - Mitsuko Kondo
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuto Sato
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan; Department of Hygiene and Public Health, Tokyo Women's Medical University, Tokyo, Japan
| | - Azusa Miyoshi
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Fumi Kobayashi
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Arimura
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Kaoru Yamashita
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoko Yanagisawa
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Etsuko Tagaya
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
9
|
Kang H, Choi Y, Kim H, Kim H, Jeong H. Sambou Bamboo salt™ down-regulates the expression levels of angiotensin-converting enzyme 2 in activated human mast cells. Food Sci Biotechnol 2024; 33:1697-1705. [PMID: 38623440 PMCID: PMC11016022 DOI: 10.1007/s10068-023-01438-3] [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: 05/17/2023] [Revised: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 04/17/2024] Open
Abstract
Mast cells have a detrimental impact on coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Sambou Bamboo salt™ (BS) suppresses mast cell-mediated inflammatory response and enhances immunity. In this study, we investigated the regulatory effects of BS on expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2) in human mast cell line (HMC)-1 cells. BS resulted in significant reductions in expression levels of ACE2 and TMPRSS2 in activated HMC-1 cells. Levels of tryptase were reduced by BS. In addition, BS blocked activation of activator protein 1 (AP-1), c-Jun NH2-terminal kinases (JNK), p38, and phosphatidylinositide-3-kinase (PI3K) in activated HMC-1 cells. Therefore, these results show that BS reduces levels of ACE2, TMPRSS2, and tryptase by inhibiting AP-1/JNK/p38/PI3K signaling pathways in mast cells. These findings can serve as valuable foundational data for the development of therapeutic agents aimed at preventing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ho‑Geun Kang
- Department of Bio-Convergence System, Graduate School, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
| | - Yu‑Jin Choi
- Department of Food Science & Technology, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
| | - Hee‑Yun Kim
- BioChip Research Center, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
| | - Hyung‑Min Kim
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, 02447 Korea
| | - Hyun‑Ja Jeong
- Department of Bio-Convergence System, Graduate School, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
- Department of Food Science & Technology, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
- BioChip Research Center, Hoseo University, 20 Hoseo-Ro, 79 Beon-Gil, Baebang-Eup, Asan, 31499 Republic of Korea
| |
Collapse
|
10
|
Lo PC, Feng JY, Hsiao YH, Su KC, Chou KT, Chen YM, Ko HK, Perng DW. Long COVID symptoms after 8-month recovery: persistent static lung hyperinflation associated with small airway dysfunction. Respir Res 2024; 25:209. [PMID: 38750527 PMCID: PMC11097537 DOI: 10.1186/s12931-024-02830-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Limited research has investigated the relationship between small airway dysfunction (SAD) and static lung hyperinflation (SLH) in patients with post-acute sequelae of COVID-19 (PASC) especially dyspnea and fatigue. METHODS 64 patients with PASC were enrolled between July 2020 and December 2022 in a prospective observational cohort. Pulmonary function tests, impulse oscillometry (IOS), and symptom questionnaires were performed two, five and eight months after acute infection. Multivariable logistic regression models were used to test the association between SLH and patient-reported outcomes. RESULTS SLH prevalence was 53.1% (34/64), irrespective of COVID-19 severity. IOS parameters and circulating CD4/CD8 T-cell ratio were significantly correlated with residual volume to total lung capacity ratio (RV/TLC). Serum CD8 + T cell count was negatively correlated with forced expiratory volume in the first second (FEV1) and forced vital capacity (FVC) with statistical significance. Of the patients who had SLH at baseline, 57% continued to have persistent SLH after eight months of recovery, with these patients tending to be older and having dyspnea and fatigue. Post-COVID dyspnea was significantly associated with SLH and IOS parameters R5-R20, and AX with adjusted odds ratios 12.4, 12.8 and 7.6 respectively. SLH was also significantly associated with fatigue. CONCLUSION SAD and a decreased serum CD4/CD8 ratio were associated with SLH in patients with PASC. SLH may persist after recovery from infection in a substantial proportion of patients. SAD and dysregulated T-cell immune response correlated with SLH may contribute to the development of dyspnea and fatigue in patients with PASC.
Collapse
Affiliation(s)
- Po-Chun Lo
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Yi-Han Hsiao
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Kang-Cheng Su
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Kun-Ta Chou
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Hsin-Kuo Ko
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC.
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC.
| | - Diahn-Warng Perng
- Department of Chest Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taipei, 11217, Taiwan, ROC.
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC.
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC.
| |
Collapse
|
11
|
Grewal T, Nguyen MKL, Buechler C. Cholesterol and COVID-19-therapeutic opportunities at the host/virus interface during cell entry. Life Sci Alliance 2024; 7:e202302453. [PMID: 38388172 PMCID: PMC10883773 DOI: 10.26508/lsa.202302453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
The rapid development of vaccines to combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections has been critical to reduce the severity of COVID-19. However, the continuous emergence of new SARS-CoV-2 subtypes highlights the need to develop additional approaches that oppose viral infections. Targeting host factors that support virus entry, replication, and propagation provide opportunities to lower SARS-CoV-2 infection rates and improve COVID-19 outcome. This includes cellular cholesterol, which is critical for viral spike proteins to capture the host machinery for SARS-CoV-2 cell entry. Once endocytosed, exit of SARS-CoV-2 from the late endosomal/lysosomal compartment occurs in a cholesterol-sensitive manner. In addition, effective release of new viral particles also requires cholesterol. Hence, cholesterol-lowering statins, proprotein convertase subtilisin/kexin type 9 antibodies, and ezetimibe have revealed potential to protect against COVID-19. In addition, pharmacological inhibition of cholesterol exiting late endosomes/lysosomes identified drug candidates, including antifungals, to block SARS-CoV-2 infection. This review describes the multiple roles of cholesterol at the cell surface and endolysosomes for SARS-CoV-2 entry and the potential of drugs targeting cholesterol homeostasis to reduce SARS-CoV-2 infectivity and COVID-19 disease severity.
Collapse
Affiliation(s)
- Thomas Grewal
- https://ror.org/0384j8v12 School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Mai Khanh Linh Nguyen
- https://ror.org/0384j8v12 School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Christa Buechler
- https://ror.org/01226dv09 Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
12
|
Owens K, Esmaeili S, Schiffer JT. Heterogeneous SARS-CoV-2 kinetics due to variable timing and intensity of immune responses. JCI Insight 2024; 9:e176286. [PMID: 38573774 PMCID: PMC11141931 DOI: 10.1172/jci.insight.176286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
The viral kinetics of documented SARS-CoV-2 infections exhibit a high degree of interindividual variability. We identified 6 distinct viral shedding patterns, which differed according to peak viral load, duration, expansion rate, and clearance rate, by clustering data from 768 infections in the National Basketball Association cohort. Omicron variant infections in previously vaccinated individuals generally led to lower cumulative shedding levels of SARS-CoV-2 than other scenarios. We then developed a mechanistic mathematical model that recapitulated 1,510 observed viral trajectories, including viral rebound and cases of reinfection. Lower peak viral loads were explained by a more rapid and sustained transition of susceptible cells to a refractory state during infection as well as by an earlier and more potent late, cytolytic immune response. Our results suggest that viral elimination occurs more rapidly during Omicron infection, following vaccination, and following reinfection due to enhanced innate and acquired immune responses. Because viral load has been linked with COVID-19 severity and transmission risk, our model provides a framework for understanding the wide range of observed SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Katherine Owens
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shadisadat Esmaeili
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Joshua T. Schiffer
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
- University of Washington, Department of Medicine, Seattle, Washington, USA
| |
Collapse
|
13
|
Gracia-Ramos AE, Ángeles-Durán GY, Flores-Gómez IR, Flores-Martínez E, Valdin-Orozco TI, Reyes-Peralta JR, Pedraza-Hervert C, Garcia-Arroyo FA, Cortés Ortíz A, Pecero-Hidalgo MJ. Standard of care plus telmisartan on respiratory failure due to COVID-19 (STAR-COVID trial). Minerva Cardiol Angiol 2024; 72:102-110. [PMID: 38231079 DOI: 10.23736/s2724-5683.23.06463-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
BACKGROUND The potential influence of renin-angiotensin inhibitors on the severity of SARS-CoV-2 infection has been considered in preclinical and observational studies with contradictory results. Therefore, we investigated the effect of telmisartan in reducing lung injury among hospitalized COVID-19 patients. METHODS The STAR-COVID trial was conducted as a prospective, parallel-group, randomized, open-label study involving hospitalized adult patients with severe COVID-19 (NCT04510662). Sixty-six patients were enrolled: 33 were assigned to the telmisartan group and 33 to the control group. The mean age of participants was 48.8 years, with 62.5% being male. Participants were randomly assigned in a 1:1 ratio to receive either telmisartan (40 mg daily for 14 days or until discharge) plus standard of care or standard of care alone. The primary outcome assessed was the initiation of mechanical ventilation within 14 days. Secondary outcomes included 30-day mortality, the need for vasopressors, hemodialysis requirements, and length of hospital stay. RESULTS Comparison between the telmisartan group and the control group revealed no significant difference in the occurrence of mechanical ventilation at 14 days (25% with telmisartan vs. 18.7% with control, P=0.579). Additionally, there were no significant differences observed in terms of mortality (25% vs. 21.9%, P=0.768), the need for vasopressors (18.8% in both groups, P=1.000), hemodialysis requirements (6.3% vs. 3.1%, P=0.500), and length of hospital stay (median of 7 days in both groups, P=0.962). CONCLUSIONS Compared with the standard of care, telmisartan therapy demonstrated no significant impact on respiratory failure in hospitalized patients with severe COVID-19.
Collapse
Affiliation(s)
- Abraham E Gracia-Ramos
- Department of Internal Medicine, La Raza Medical Complex, Mexican Social Security Institute, UMAE Hospital General "Dr. Gaudencio González Garza, " Mexico City, Mexico -
- School of Medicine, Section of Graduate Studies and Research, National Polytechnic Institute of Mexico, Mexico City, Mexico -
| | - Gissel Y Ángeles-Durán
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Iván R Flores-Gómez
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Emmanuel Flores-Martínez
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Tania I Valdin-Orozco
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Jesús R Reyes-Peralta
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - César Pedraza-Hervert
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Flor A Garcia-Arroyo
- Department of Internal Medicine, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - Antonio Cortés Ortíz
- Department of Cardiology, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| | - María J Pecero-Hidalgo
- Director Office, Hospital Regional de Alta Especialidad de Zumpango (HRAEZ), Zumpango, Mexico
| |
Collapse
|
14
|
Nikiforuk AM, Kuchinski KS, Short K, Roman S, Irvine MA, Prystajecky N, Jassem AN, Patrick DM, Sekirov I. Nasopharyngeal angiotensin converting enzyme 2 (ACE2) expression as a risk-factor for SARS-CoV-2 transmission in concurrent hospital associated outbreaks. BMC Infect Dis 2024; 24:262. [PMID: 38408924 PMCID: PMC10898082 DOI: 10.1186/s12879-024-09067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/28/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Widespread human-to-human transmission of the severe acute respiratory syndrome coronavirus two (SARS-CoV-2) stems from a strong affinity for the cellular receptor angiotensin converting enzyme two (ACE2). We investigate the relationship between a patient's nasopharyngeal ACE2 transcription and secondary transmission within a series of concurrent hospital associated SARS-CoV-2 outbreaks in British Columbia, Canada. METHODS Epidemiological case data from the outbreak investigations was merged with public health laboratory records and viral lineage calls, from whole genome sequencing, to reconstruct the concurrent outbreaks using infection tracing transmission network analysis. ACE2 transcription and RNA viral load were measured by quantitative real-time polymerase chain reaction. The transmission network was resolved to calculate the number of potential secondary cases. Bivariate and multivariable analyses using Poisson and Negative Binomial regression models was performed to estimate the association between ACE2 transcription the number of SARS-CoV-2 secondary cases. RESULTS The infection tracing transmission network provided n = 76 potential transmission events across n = 103 cases. Bivariate comparisons found that on average ACE2 transcription did not differ between patients and healthcare workers (P = 0.86). High ACE2 transcription was observed in 98.6% of transmission events, either the primary or secondary case had above average ACE2. Multivariable analysis found that the association between ACE2 transcription (log2 fold-change) and the number of secondary transmission events differs between patients and healthcare workers. In health care workers Negative Binomial regression estimated that a one-unit change in ACE2 transcription decreases the number of secondary cases (β = -0.132 (95%CI: -0.255 to -0.0181) adjusting for RNA viral load. Conversely, in patients a one-unit change in ACE2 transcription increases the number of secondary cases (β = 0.187 (95% CI: 0.0101 to 0.370) adjusting for RNA viral load. Sensitivity analysis found no significant relationship between ACE2 and secondary transmission in health care workers and confirmed the positive association among patients. CONCLUSION Our study suggests that ACE2 transcription has a positive association with SARS-CoV-2 secondary transmission in admitted inpatients, but not health care workers in concurrent hospital associated outbreaks, and it should be further investigated as a risk-factor for viral transmission.
Collapse
Affiliation(s)
- Aidan M Nikiforuk
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada
| | - Kevin S Kuchinski
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada
| | - Katy Short
- Fraser Health Authority, V3L 3C2, New Westminster, BC, Canada
| | - Susan Roman
- Fraser Health Authority, V3L 3C2, New Westminster, BC, Canada
| | - Mike A Irvine
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, V5A 1S6, Burnaby, BC, Canada
| | - Natalie Prystajecky
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada
| | - Agatha N Jassem
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada
| | - David M Patrick
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada
| | - Inna Sekirov
- British Columbia Centre for Disease Control, V5Z 4R4, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, V6T 1Z4, Vancouver, BC, Canada.
| |
Collapse
|
15
|
Owens K, Esmaeili-Wellman S, Schiffer JT. Heterogeneous SARS-CoV-2 kinetics due to variable timing and intensity of immune responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.20.23294350. [PMID: 37662228 PMCID: PMC10473815 DOI: 10.1101/2023.08.20.23294350] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The viral kinetics of documented SARS-CoV-2 infections exhibit a high degree of inter-individual variability. We identified six distinct viral shedding patterns, which differed according to peak viral load, duration, expansion rate and clearance rate, by clustering data from 768 infections in the National Basketball Association cohort. Omicron variant infections in previously vaccinated individuals generally led to lower cumulative shedding levels of SARS-CoV-2 than other scenarios. We then developed a mechanistic mathematical model that recapitulated 1510 observed viral trajectories, including viral rebound and cases of reinfection. Lower peak viral loads were explained by a more rapid and sustained transition of susceptible cells to a refractory state during infection, as well as an earlier and more potent late, cytolytic immune response. Our results suggest that viral elimination occurs more rapidly during omicron infection, following vaccination, and following re-infection due to enhanced innate and acquired immune responses. Because viral load has been linked with COVID-19 severity and transmission risk, our model provides a framework for understanding the wide range of observed SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Katherine Owens
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division
| | | | - Joshua T Schiffer
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division
- University of Washington, Department of Medicine
| |
Collapse
|
16
|
Carossino M, Izadmehr S, Trujillo JD, Gaudreault NN, Dittmar W, Morozov I, Balasuriya UBR, Cordon-Cardo C, García-Sastre A, Richt JA. ACE2 and TMPRSS2 distribution in the respiratory tract of different animal species and its correlation with SARS-CoV-2 tissue tropism. Microbiol Spectr 2024; 12:e0327023. [PMID: 38230954 PMCID: PMC10846196 DOI: 10.1128/spectrum.03270-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024] Open
Abstract
A wide range of animal species show variable susceptibility to SARS-CoV-2; however, host factors associated with varied susceptibility remain to be defined. Here, we examined whether susceptibility to SARS-CoV-2 and virus tropism in different animal species are dependent on the expression and distribution of the virus receptor angiotensin-converting enzyme 2 (ACE2) and the host cell factor transmembrane serine protease 2 (TMPRSS2). We cataloged the upper and lower respiratory tract of multiple animal species and humans in a tissue-specific manner and quantitatively evaluated the distribution and abundance of ACE2 and TMPRSS2 mRNA in situ. Our results show that: (i) ACE2 and TMPRSS2 mRNA are abundant in the conduction portion of the respiratory tract, (ii) ACE2 mRNA occurs at a lower abundance compared to TMPRSS2 mRNA, (iii) co-expression of ACE2-TMPRSS2 mRNAs is highest in those species with the highest susceptibility to SARS-CoV-2 infection (i.e., cats, Syrian hamsters, and white-tailed deer), and (iv) expression of ACE2 and TMPRSS2 mRNA was not altered following SARS-CoV-2 infection. Our results demonstrate that while specific regions of the respiratory tract are enriched in ACE2 and TMPRSS2 mRNAs in different animal species, this is only a partial determinant of susceptibility to SARS-CoV-2 infection.IMPORTANCESARS-CoV-2 infects a wide array of domestic and wild animals, raising concerns regarding its evolutionary dynamics in animals and potential for spillback transmission of emerging variants to humans. Hence, SARS-CoV-2 infection in animals has significant public health relevance. Host factors determining animal susceptibility to SARS-CoV-2 are vastly unknown, and their characterization is critical to further understand susceptibility and viral dynamics in animal populations and anticipate potential spillback transmission. Here, we quantitatively assessed the distribution and abundance of the two most important host factors, angiotensin-converting enzyme 2 and transmembrane serine protease 2, in the respiratory tract of various animal species and humans. Our results demonstrate that while specific regions of the respiratory tract are enriched in these two host factors, they are only partial determinants of susceptibility. Detailed analysis of additional host factors is critical for our understanding of the underlying mechanisms governing viral susceptibility and reservoir hosts.
Collapse
Affiliation(s)
- Mariano Carossino
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Sudeh Izadmehr
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessie D. Trujillo
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Wellesley Dittmar
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adolfo García-Sastre
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
17
|
Cha MJ, Solomon JJ, Lee JE, Choi H, Chae KJ, Lee KS, Lynch DA. Chronic Lung Injury after COVID-19 Pneumonia: Clinical, Radiologic, and Histopathologic Perspectives. Radiology 2024; 310:e231643. [PMID: 38193836 PMCID: PMC10831480 DOI: 10.1148/radiol.231643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/06/2023] [Accepted: 09/26/2023] [Indexed: 01/10/2024]
Abstract
With the COVID-19 pandemic having lasted more than 3 years, concerns are growing about prolonged symptoms and respiratory complications in COVID-19 survivors, collectively termed post-COVID-19 condition (PCC). Up to 50% of patients have residual symptoms and physiologic impairment, particularly dyspnea and reduced diffusion capacity. Studies have also shown that 24%-54% of patients hospitalized during the 1st year of the pandemic exhibit radiologic abnormalities, such as ground-glass opacity, reticular opacity, bronchial dilatation, and air trapping, when imaged more than 1 year after infection. In patients with persistent respiratory symptoms but normal results at chest CT, dual-energy contrast-enhanced CT, xenon 129 MRI, and low-field-strength MRI were reported to show abnormal ventilation and/or perfusion, suggesting that some lung injury may not be detectable with standard CT. Histologic patterns in post-COVID-19 lung disease include fibrosis, organizing pneumonia, and vascular abnormality, indicating that different pathologic mechanisms may contribute to PCC. Therefore, a comprehensive imaging approach is necessary to evaluate and diagnose patients with persistent post-COVID-19 symptoms. This review will focus on the long-term findings of clinical and radiologic abnormalities and describe histopathologic perspectives. It also addresses advanced imaging techniques and deep learning approaches that can be applied to COVID-19 survivors. This field remains an active area of research, and further follow-up studies are warranted for a better understanding of the chronic stage of the disease and developing a multidisciplinary approach for patient management.
Collapse
Affiliation(s)
- Min Jae Cha
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - Joshua J. Solomon
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - Jong Eun Lee
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - Hyewon Choi
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - Kum Ju Chae
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - Kyung Soo Lee
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| | - David A. Lynch
- From the Department of Radiology, Chung-Ang University Hospital,
Seoul, Korea (M.J.C., H.C.); Departments of Medicine (J.J.S.) and Radiology
(K.J.C., D.A.L.), National Jewish Health, 1400 Jackson St, Denver, CO 80206;
Department of Radiology, Chonnam National University Hospital, Gwangju, Republic
of Korea (J.E.L.); Department of Radiology, Research Institute of Clinical
Medicine of Jeonbuk National University, Biomedical Research Institute of
Jeonbuk National University Hospital, Jeonju, Republic of Korea (K.J.C); and
Department of Radiology, Sungkyunkwan University School of Medicine and Samsung
ChangWon Hospital, Gyeongsangnam, Republic of Korea (K.S.L.)
| |
Collapse
|
18
|
Dos Santos JF, Dos Reis EM, Berti FV, Colla G, Koepp J, Nunes VA. CALU-3 lung cells three-dimensionally assembled onto CellFate® matrix present angiotensin-converting enzyme-2 activity. Biotechnol Bioeng 2023; 120:3602-3611. [PMID: 37691178 DOI: 10.1002/bit.28552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023]
Abstract
Currently, there is a great need for the development of three-dimensional (3D) in vitro lung models. Particularly, the production of a suitable 3D model of pulmonary epithelium for understanding the pathophysiology of diseases such as the COVID-19 must consider the tissue architecture and presence, for example, of the angiotensin-converting enzyme-2 (ACE-2) in the cells. Different polymeric membranes are being used to support cell culturing, especially of lung cells, however, there is still no information about the culture of these cells onto bacterial nanocellulose (BNC) matrices. We have used the BNC matrix CellFate® as a support for the assembly of a 3D in vitro model of lung epithelium, composed of human lung fibroblasts (HLF) and lung adenocarcinoma cells (CALU-3). CellFate® matrices were made from bacterial fermentation resulting in a natural and biocompatible biopolymer. Cells were cultured onto CellFate® and maintained in a 5% CO2 humidified atmosphere at 37°C. Cell viability was assessed by the resazurin method The samples were, then, exposed to the air-liquid interface (ALI), and histologically analyzed. ACE-2 activity was verified on the hydrolyze of the fluorogenic substrate Mca-APK(Dnp)-OH, and its presence was evaluated by flow cytometry. The expression of the anionic transporter SLCO3A1 was evaluated by qPCR. Cell viability analysis indicates that CellFate® was not toxic to these cells. By flow cytometry, the presence of the ACE-2 was identified in the CALU-3 cells surface corroborating the results obtained from enzymatic activity analysis. The SLCO3A1 transporter expression was identified in cells cultured onto CellFate®, but not in cells cultured onto the transwell (control). CALU-3 cells cultivated onto CellFate® resulted in a pseudostratified organization, a typical morphology of the human respiratory tract epithelium. The current model opens perspectives for studies involving physiological characterization, improving its relevance for the understanding of the pathophysiology of diseases as well as the response to drugs.
Collapse
Affiliation(s)
- Jeniffer Farias Dos Santos
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities (EACH) of University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | - Janice Koepp
- Biocelltis Biotechnology SA, Florianópolis, Brazil
| | - Viviane Abreu Nunes
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities (EACH) of University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
19
|
Cai J, Ma W, Wang X, Chang H, Wei Z, Li J, Zeng M. The spike protein of SARS-CoV-2 induces inflammation and EMT of lung epithelial cells and fibroblasts through the upregulation of GADD45A. Open Med (Wars) 2023; 18:20230779. [PMID: 38025528 PMCID: PMC10656760 DOI: 10.1515/med-2023-0779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 06/27/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023] Open
Abstract
Lung epithelial cells and fibroblasts poorly express angiotensin-converting enzyme 2, and the study aimed to investigate the role of the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on inflammation and epithelial-mesenchymal transition (EMT) in two lung cell lines and to understand the potential mechanism. Lung epithelial cells (BEAS-2B) and fibroblasts (MRC-5) were treated with the spike protein, then inflammatory and EMT phenotypes were detected by enzyme-linked immunosorbent assay, Transwell, and western blot assays. RNA-sequence and bioinformatic analyses were performed to identify dysregulated genes. The roles of the candidate genes were further investigated. The results showed that treatment with 1,000 ng/mL of spike protein in two lung cell lines caused increased levels of IL-6, TNF-α, CXCL1, and CXCL3, and the occurrence of EMT. RNA-sequence identified 4,238 dysregulated genes in the spike group, and 18 candidate genes were involved in both inflammation- and EMT-related processes. GADD45A had the highest verified fold change (abs), and overexpression of GADD45A promoted the secretion of cytokines and EMT in the two lung cell lines. In conclusion, the spike protein induces inflammation and EMT in lung epithelial cells and fibroblasts by upregulating GADD45A, providing a new target to inhibit inflammation and EMT.
Collapse
Affiliation(s)
- Jiehao Cai
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Wenjie Ma
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Xiangshi Wang
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Hailing Chang
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Zhongqiu Wei
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Jingjing Li
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| | - Mei Zeng
- Department of Infectious Diseases, Children’s Hospital of Fudan University, Shanghai201102, China
| |
Collapse
|
20
|
Adimulam T, Arumugam T, Naidoo A, Naidoo K, Ramsuran V. Polymorphisms within the SARS-CoV-2 Human Receptor Genes Associate with Variable Disease Outcomes across Ethnicities. Genes (Basel) 2023; 14:1798. [PMID: 37761938 PMCID: PMC10531089 DOI: 10.3390/genes14091798] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The contribution of human genes to the variability of disease outcomes has been shown to be important across infectious diseases. Studies have shown mutations within specific human genes are associated with variable COVID-19 outcomes. We focused on the SARS-CoV-2 receptors/co-receptors to identify the role of specific polymorphisms within ACE2, TMPRSS2, NRP1 and CD147. Polymorphisms within ACE2 (rs2285666), TMPRSS2 (rs12329760), CD147 (rs8259) and NRP1 (rs10080) have been shown to associate with COVID-19 severity. Using cryopreserved samples from COVID-19-positive African, European and South Asian individuals within South Africa, we determined genotype frequencies. The genetic variant rs2285666 was associated with COVID-19 severity with an ethnic bias. African individuals with a CC genotype demonstrate more severe COVID-19 outcomes (OR = 7.5; 95% CI 1.164-80.89; p = 0.024) compared with those with a TT genotype. The expressions of ACE2 and SARS-CoV-2 viral load were measured using droplet digital PCR. Our results demonstrate rs2285666 and rs10080 were significantly associated with increased SARS-CoV-2 viral load and worse outcomes in certain ethnicities. This study demonstrates two important findings. Firstly, SARS-CoV-2 viral load is significantly lower in Africans compared with individuals of European and South Asian descent (p = 0.0002 and p < 0.0001). Secondly, SARS-CoV-2 viral load associates with specific SARS-CoV-2 receptor variants. A limited number of studies have examined the receptor/co-receptor genes within Africa. This study investigated genetic variants within the SARS-CoV-2 receptor/co-receptor genes and their association with COVID-19 severity and SARS-CoV-2 viral load across different ethnicities. We provide a genetic basis for differences in COVID-19 severity across ethnic groups in South Africa, further highlighting the importance of further investigation to determine potential therapeutic targets and to guide vaccination strategies that may prioritize specific genotypes.
Collapse
Affiliation(s)
- Theolan Adimulam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa; (T.A.); (T.A.)
| | - Thilona Arumugam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa; (T.A.); (T.A.)
| | - Anushka Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (A.N.); (K.N.)
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (A.N.); (K.N.)
- South African Medical Research Council (SAMRC), Durban 4013, South Africa
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa; (T.A.); (T.A.)
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (A.N.); (K.N.)
| |
Collapse
|
21
|
Wu SC, Arthur CM, Jan HM, Garcia-Beltran WF, Patel KR, Rathgeber MF, Verkerke HP, Cheedarla N, Jajosky RP, Paul A, Neish AS, Roback JD, Josephson CD, Wesemann DR, Kalman D, Rakoff-Nahoum S, Cummings RD, Stowell SR. Blood group A enhances SARS-CoV-2 infection. Blood 2023; 142:742-747. [PMID: 37367252 PMCID: PMC10294591 DOI: 10.1182/blood.2022018903] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/18/2023] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
Among the risk factors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), ABO(H) blood group antigens are among the most recognized predictors of infection. However, the mechanisms by which ABO(H) antigens influence susceptibility to COVID-19 remain incompletely understood. The receptor-binding domain (RBD) of SARS-CoV-2, which facilitates host cell engagement, bears significant similarity to galectins, an ancient family of carbohydrate-binding proteins. Because ABO(H) blood group antigens are carbohydrates, we compared the glycan-binding specificity of SARS-CoV-2 RBD with that of galectins. Similar to the binding profile of several galectins, the RBDs of SARS-CoV-2, including Delta and Omicron variants, exhibited specificity for blood group A. Not only did each RBD recognize blood group A in a glycan array format, but each SARS-CoV-2 virus also displayed a preferential ability to infect blood group A-expressing cells. Preincubation of blood group A cells with a blood group-binding galectin specifically inhibited the blood group A enhancement of SARS-CoV-2 infection, whereas similar incubation with a galectin that does not recognize blood group antigens failed to impact SARS-CoV-2 infection. These results demonstrated that SARS-CoV-2 can engage blood group A, providing a direct link between ABO(H) blood group expression and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shang-Chuen Wu
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Connie M Arthur
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hau-Ming Jan
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Wilfredo F Garcia-Beltran
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA
| | - Kashyap R Patel
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Matthew F Rathgeber
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hans P Verkerke
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Narayanaiah Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Ryan Philip Jajosky
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anu Paul
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - John D Roback
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA
| | - Cassandra D Josephson
- Department of Hematology and Oncology, Johns Hopkins University All Children's Hospital, St. Petersburg, FL
| | - Duane R Wesemann
- Division of Allergy and Clinical Immunology and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daniel Kalman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Seth Rakoff-Nahoum
- Division of Infectious Disease, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Richard D Cummings
- National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Sean R Stowell
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Razi O, Teixeira AM, Tartibian B, Zamani N, Knechtle B. Respiratory issues in patients with multiple sclerosis as a risk factor during SARS-CoV-2 infection: a potential role for exercise. Mol Cell Biochem 2023; 478:1533-1559. [PMID: 36411399 PMCID: PMC9684932 DOI: 10.1007/s11010-022-04610-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease-2019 (COVID-19) is associated with cytokine storm and is characterized by acute respiratory distress syndrome (ARDS) and pneumonia problems. The respiratory system is a place of inappropriate activation of the immune system in people with multiple sclerosis (MS), and this may cause damage to the lung and worsen both MS and infections.The concerns for patients with multiple sclerosis are because of an enhance risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The MS patients pose challenges in this pandemic situation, because of the regulatory defect of autoreactivity of the immune system and neurological and respiratory tract symptoms. In this review, we first indicate respiratory issues associated with both diseases. Then, the main mechanisms inducing lung damages and also impairing the respiratory muscles in individuals with both diseases is discussed. At the end, the leading role of physical exercise on mitigating respiratory issues inducing mechanisms is meticulously evaluated.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Ana Maria Teixeira
- Research Center for Sport and Physical Activity, Faculty of Sport Sciences and Physical Education, University of Coimbra, Coimbra, Portugal
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, 9001 St. Gallen, Switzerland
| |
Collapse
|
23
|
Ma X, Liu L, Sun J, Zhang Y, Liu Y, Liu M, Wang B, Chang Y, Han Y, Gai Z. Expression of circulating angiotensin-converting enzyme 2 in children with asthma and the effects of inhaled corticosteroids. Immun Inflamm Dis 2023; 11:e922. [PMID: 37506151 PMCID: PMC10373566 DOI: 10.1002/iid3.922] [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: 02/10/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The global spread of coronavirus disease 2019 (COVID-19) has resulted in a significant disease burden, yet asthma patients do not have the expected high morbidity and mortality rates in the pandemics of COVID-19. OBJECTIVE To find the difference of angiotensin-converting enzyme 2 (ACE2) in asthma and nonasthma children and evaluate the effect of inhaled corticosteroids (ICS) on its expression. METHODS The ACE2, immunoglobulin E (IgE), and eosinophils were tested in different children. RESULTS A total of 157 children aged 3-16 years were enrolled. The expression of ACE2 in asthma children were lower than nonasthma children (T = -2.512, p = .013). Allergic nonasthma children had a significant higher ACE2 expression than children with allergic asthma (p = .013) and nonallergic asthma (p = .029). The expression of ACE2 had no significant difference between first-diagnosed asthma children and that had been treated with ICS for ≥6 months (F = 0.028, p = .598). The allergic asthma children showed a significantly higher eosinophils cells (EC) count than the allergic nonasthma (W = 200, p < .001) and nonallergic nonasthma children (W = 1089, p < .001). Nonallergic asthma children also had a significant higher EC count than the allergic non-asthma (W = 182.5, p < .001) and nonallergic non-asthma (W = 200.5, p < .001) children. There was no significant difference in IgE levels between asthmatic children and non-asthmatic children (W = 2792.5, p = .18). CONCLUSION Circulating ACE2 levels in asthmatic children were lower than those in non-asthmatic children and ICS treatment for ≥6 months did not affect the expression of ACE2 in peripheral blood in the asthma children.
Collapse
Affiliation(s)
- Xiang Ma
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Lesen Liu
- Department of Infectious Diseases, Huaiyin People's Hospital, Jinan, China
| | - Jing Sun
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Yun Zhang
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Yanqin Liu
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Miao Liu
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Bing Wang
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Yuna Chang
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| | - Yuling Han
- Department of Respiratory Diseases, Children's Hospital affiliated to Shandong University, Jinan, China
| | - Zhongtao Gai
- Jinan Key Lab of Respiratory Diseases for Children, Jinan Children's Hospital, Jinan, China
| |
Collapse
|
24
|
Meyerholz DK, Leidinger MR, Adam Goeken J, Businga TR, Vizuett S, Akers A, Evans I, Zhang Y, Engelhardt JF. Immunohistochemical detection of MUC5AC and MUC5B mucins in ferrets. BMC Res Notes 2023; 16:111. [PMID: 37349833 PMCID: PMC10286488 DOI: 10.1186/s13104-023-06388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE Cystic fibrosis (CF) is a genetic condition that causes abnormal mucus secretions in affected organs. MUC5AC and MUC5B are gel-forming mucins and frequent targets for investigations in CF tissues. Our objective was to qualify MUC5AC and MUC5B immunohistochemical techniques to provide a useful tool to identify, localize and interpret mucin expression in ferret tissues. RESULTS MUC5AC and MUC5B mucins were detected most commonly in large airways and least in small airways, consistent with reported goblet cell density in airway surface epithelia. We evaluated whether staining method affected the detection of goblet cell mucins in serial sections of bronchial surface epithelia. Significant differences between stains were not observed suggesting common co-expression MUC5AC and MUC5B proteins in goblet cells of airway surface epithelia. Gallbladder and stomach tissues are reported to have differential mucin enrichment, so we tested these tissues in wildtype ferrets. Stomach tissues were enriched in MUC5AC and gallbladder tissues enriched in MUC5B, mucin enrichment similar to human tissues. Mucin immunostaining techniques were further qualified for specificity using lung tissue from recently generated MUC5AC-/- and MUC5B-/- ferrets. Qualified techniques for MUC5AC and MUC5B immunohistochemistry will be useful tools for mucin tissue studies in CF and other ferret models.
Collapse
Affiliation(s)
- David K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Mariah R. Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - J. Adam Goeken
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Thomas R. Businga
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Sebastian Vizuett
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Allison Akers
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Idil Evans
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Yan Zhang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
25
|
Sharifi E, Yousefiasl S, Trovato M, Sartorius R, Esmaeili Y, Goodarzi H, Ghomi M, Bigham A, Moghaddam FD, Heidarifard M, Pourmotabed S, Nazarzadeh Zare E, Paiva-Santos AC, Rabiee N, Wang X, Tay FR. Nanostructures for prevention, diagnosis, and treatment of viral respiratory infections: from influenza virus to SARS-CoV-2 variants. J Nanobiotechnology 2023; 21:199. [PMID: 37344894 PMCID: PMC10283343 DOI: 10.1186/s12951-023-01938-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/24/2023] [Indexed: 06/23/2023] Open
Abstract
Viruses are a major cause of mortality and socio-economic downfall despite the plethora of biopharmaceuticals designed for their eradication. Conventional antiviral therapies are often ineffective. Live-attenuated vaccines can pose a safety risk due to the possibility of pathogen reversion, whereas inactivated viral vaccines and subunit vaccines do not generate robust and sustained immune responses. Recent studies have demonstrated the potential of strategies that combine nanotechnology concepts with the diagnosis, prevention, and treatment of viral infectious diseases. The present review provides a comprehensive introduction to the different strains of viruses involved in respiratory diseases and presents an overview of recent advances in the diagnosis and treatment of viral infections based on nanotechnology concepts and applications. Discussions in diagnostic/therapeutic nanotechnology-based approaches will be focused on H1N1 influenza, respiratory syncytial virus, human parainfluenza virus type 3 infections, as well as COVID-19 infections caused by the SARS-CoV-2 virus Delta variant and new emerging Omicron variant.
Collapse
Affiliation(s)
- Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran.
| | - Satar Yousefiasl
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Yasaman Esmaeili
- School of Advanced Technologies in Medicine, Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Hamid Goodarzi
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran
| | - Ashkan Bigham
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133, Rome, Italy
| | - Maryam Heidarifard
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Samiramis Pourmotabed
- Department of Emergency Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | | | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
26
|
Beyer D, Vaccarin C, Deupi X, Mapanao AK, Cohrs S, Sozzi-Guo F, Grundler PV, van der Meulen NP, Wang J, Tanriver M, Bode JW, Schibli R, Müller C. A tool for nuclear imaging of the SARS-CoV-2 entry receptor: molecular model and preclinical development of ACE2-selective radiopeptides. EJNMMI Res 2023; 13:32. [PMID: 37074529 PMCID: PMC10113987 DOI: 10.1186/s13550-023-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/29/2023] [Indexed: 04/20/2023] Open
Abstract
PURPOSE The angiotensin converting enzyme-2 (ACE2)-entry receptor of SARS-CoV-2-and its homologue, the angiotensin-converting enzyme (ACE), play a pivotal role in maintaining cardiovascular homeostasis. Potential changes in ACE2 expression levels and dynamics after SARS-CoV-2 infection have been barely investigated. The aim of this study was to develop an ACE2-targeting imaging agent as a noninvasive imaging tool to determine ACE2 regulation. METHODS DOTA-DX600, NODAGA-DX600 and HBED-CC-DX600 were obtained through custom synthesis and labeled with gallium-67 (T1/2 = 3.26 d) as a surrogate radioisotope for gallium-68 (T1/2 = 68 min). ACE2- and ACE-transfected HEK cells were used for the in vitro evaluation of these radiopeptides. The in vivo tissue distribution profiles of the radiopeptides were assessed in HEK-ACE2 and HEK-ACE xenografted mice and imaging studies were performed using SPECT/CT. RESULTS The highest molar activity was obtained for [67Ga]Ga-HBED-CC-DX600 (60 MBq/nmol), whereas the labeling efficiency of the other peptides was considerably lower (20 MBq/nmol). The radiopeptides were stable over 24 h in saline (> 99% intact peptide). All radiopeptides showed uptake in HEK-ACE2 cells (36-43%) with moderate ACE2-binding affinity (KD value: 83-113 nM), but no uptake in HEK-ACE cells (< 0.1%) was observed. Accumulation of the radiopeptides was observed in HEK-ACE2 xenografts (11-16% IA/g) at 3 h after injection, but only background signals were seen in HEK-ACE xenografts (< 0.5% IA/g). Renal retention was still high 3 h after injection of [67Ga]Ga-DOTA-DX600 and [67Ga]Ga-NODAGA-DX600 (~ 24% IA/g), but much lower for [67Ga]Ga-HBED-CC-DX600 (7.2 ± 2.2% IA/g). SPECT/CT imaging studies confirmed the most favorable target-to-nontarget ratio for [67Ga]Ga-HBED-CC-DX600. CONCLUSIONS This study demonstrated ACE2 selectivity for all radiopeptides. [67Ga]Ga-HBED-CC-DX600 was revealed as the most promising candidate due to its favorable tissue distribution profile. Importantly, the HBED-CC chelator enabled 67Ga-labeling at high molar activity, which would be essential to obtain images with high signal-to-background contrast to detect (patho)physiological ACE2 expression levels in patients.
Collapse
Affiliation(s)
- Darja Beyer
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Christian Vaccarin
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Xavier Deupi
- Condensed Matter Theory Group, Division of Scientific Computing, Theory, and Data, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Susan Cohrs
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Fan Sozzi-Guo
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Jinling Wang
- Institute of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Matthias Tanriver
- Institute of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Jeffrey W Bode
- Institute of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland.
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland.
| |
Collapse
|
27
|
Saleem W, Ren X, Van Den Broeck W, Nauwynck H. Changes in intestinal morphology, number of mucus-producing cells and expression of coronavirus receptors APN, DPP4, ACE2 and TMPRSS2 in pigs with aging. Vet Res 2023; 54:34. [PMID: 37055856 PMCID: PMC10100624 DOI: 10.1186/s13567-023-01169-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/01/2023] [Indexed: 04/15/2023] Open
Abstract
Porcine enteric viral infections cause high morbidity and mortality in young piglets (<3 weeks). Later, these rates decrease with age. This age-dependent infectivity remains largely unexplored. This study investigated the changes in intestinal morphology, number of mucus-producing cells and expression level of coronavirus receptors in three age groups of pigs. Villus height and crypt depth increased with age from 3 days to 3 months in duodenum and ileum but not in mid-jejunum, where the villus height decreased from 580 µm at 3 days to 430 µm at 3 months. Enterocyte length-to-width ratio increased from 3 days to 3 months in all intestinal regions. The number of mucus-producing cells increased with age in the intestinal villi and crypts. The Brunner's glands of the duodenum contained the highest concentration of mucus-producing cells. The expression of coronavirus receptor APN was highest in the small intestinal villi at all ages. DPP4 expression slightly decreased over time in jejunum and ileum; it was highest in the ileal villi of 3-day-old piglets (70.2% of cells). ACE2 and TMPRSS2 positive cells increased with age in jejunal and ileal crypts and were particularly dominant in the ileal crypts (> 45% of cells). Except for the expression of DPP4 in the jejunum and ileum of young pigs, the expression pattern of the selected coronavirus receptors was very different and not correlated with the age-dependent susceptibility to viral infections. In contrast, the number of mucus-producing cells increased over time and may play an essential role in protecting enteric mucosae against intestinal viruses.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| | - Xiaolei Ren
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| |
Collapse
|
28
|
Ziablitsev DS, Kozyk M, Strubchevska K, Dyadyk OO, Ziablitsev SV. Lung Expression of Macrophage Markers CD68 and CD163, Angiotensin Converting Enzyme 2 (ACE2), and Caspase-3 in COVID-19. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040714. [PMID: 37109672 PMCID: PMC10144424 DOI: 10.3390/medicina59040714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023]
Abstract
Background and Objectives: The coronavirus (SARS-CoV-2) damages all systems and organs. Yet, to a greater extent, the lungs are particularly involved, due to the formation of diffuse exudative inflammation in the form of acute respiratory distress syndrome (ARDS) with next progression to pulmonary fibrosis. SARS-associated lung damage is accompanied by the pronounced activation of mononuclear cells, damage of the alveoli and microvessels, and the development of organized pneumonia. To study the expression of macrophage markers (CD68 and CD163), angiotensin-converting enzyme-2 (ACE2), and caspase-3 on the results of two fatal clinical observations of COVID-19. Materials and Methods: In both clinical cases, the female patients died from complications of confirmed COVID-19. Conventional morphological and immunohistochemical methods were used. Results: There was an acute exudative hemorrhagic pneumonia with the formation of hyaline membranes, focal organization of fibrin, stromal sclerosis, stasis, and thrombus formation in the lung vessels. Signs such as the formation of hyaline membranes, organization, and fibrosis were more pronounced in severe disease activity. The activation of CD68+/CD163+ macrophages could cause cell damage at an early stage of pneumonia development, and subsequently cause fibrotic changes in lung tissue. ACE2 expression in lung tissue was not detected in severe pneumonia, while in moderate pneumonia, weak expression was noted in individual cells of the alveolar epithelium and vascular endothelium. Conclusions: This finding could show the dependence of ACE2 expression on the severity of the inflammatory process in the lungs. The expression of caspase-3 was more pronounced in severe pneumonia.
Collapse
Affiliation(s)
- Denis S Ziablitsev
- Department of Pathophysiology, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| | - Marko Kozyk
- Department of Internal Medicine, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48073, USA
| | - Kateryna Strubchevska
- Department of Internal Medicine, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48073, USA
| | - Olena O Dyadyk
- Department of Pathologic and Topographic Anatomy, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine
| | - Sergiy V Ziablitsev
- Department of Pathophysiology, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| |
Collapse
|
29
|
Benfante A, Pirrello G, Sala F, Seminara G, Scichilone N. Coronavirus disease 2019 and severe asthma. Curr Opin Allergy Clin Immunol 2023; 23:193-198. [PMID: 36752375 DOI: 10.1097/aci.0000000000000893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
PURPOSE OF REVIEW The relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the most severe forms of asthma has been an object of discussion. Indeed, it is not clear whether asthma is among the risk factors for the occurrence of severe coronavirus disease 2019 (COVID-19) disease, or rather it plays a protective role against the worsening of the respiratory involvement in the SARS-CoV-2 infection. On the other hand, the extent to which coronavirus infection may trigger asthma attacks is still partly unknown. The current investigation aims at reviewing the available literature on the topic to address factors influencing this relationship. RECENT FINDINGS Based on recent observations, it is likely that type 2 inflammation plays a protective role against SARS-CoV-2 infection and disease. In particular, asthmatics show different expression of angiotensin-converting enzyme2 (ACE2) and Transmembrane protease, serine 2 (TMPRSS2) that are responsible for a reduced risk of infection as well as lower risk of hospitalization. Interestingly, studies showed a safe profile of inhaled corticosteroids and biological drugs in SARS-CoV-2 infection. In addition, inhaled corticosteroid could play a protective role against worsening of asthma. SUMMARY The current findings suggest that current treatment for asthma should be maintained to avoid severe exacerbations. Severe asthmatics under biological treatment should continue their medications, and be encouraged to receive COVID-19 vaccines.
Collapse
Affiliation(s)
- Alida Benfante
- Dipartimento Universitario di Promozione della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Division of Respiratory Medicine, "Paolo Giaccone" University Hospital, University of Palermo, Palermo, Italy
| | | | | | | | | |
Collapse
|
30
|
Devaux CA, Lagier JC. Unraveling the Underlying Molecular Mechanism of 'Silent Hypoxia' in COVID-19 Patients Suggests a Central Role for Angiotensin II Modulation of the AT1R-Hypoxia-Inducible Factor Signaling Pathway. J Clin Med 2023; 12:jcm12062445. [PMID: 36983445 PMCID: PMC10056466 DOI: 10.3390/jcm12062445] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
A few days after being infected with SARS-CoV-2, a fraction of people remain asymptomatic but suffer from a decrease in arterial oxygen saturation in the absence of apparent dyspnea. In light of our clinical investigation on the modulation of molecules belonging to the renin angiotensin system (RAS) in COVID-19 patients, we propose a model that explains 'silent hypoxia'. The RAS imbalance caused by SARS-CoV-2 results in an accumulation of angiotensin 2 (Ang II), which activates the angiotensin 2 type 1 receptor (AT1R) and triggers a harmful cascade of intracellular signals leading to the nuclear translocation of the hypoxia-inducible factor (HIF)-1α. HIF-1α transactivates many genes including the angiotensin-converting enzyme 1 (ACE1), while at the same time, ACE2 is downregulated. A growing number of cells is maintained in a hypoxic condition that is self-sustained by the presence of the virus and the ACE1/ACE2 ratio imbalance. This is associated with a progressive worsening of the patient's biological parameters including decreased oxygen saturation, without further clinical manifestations. When too many cells activate the Ang II-AT1R-HIF-1α axis, there is a 'hypoxic spillover', which marks the tipping point between 'silent' and symptomatic hypoxia in the patient. Immediate ventilation is required to prevent the 'hypoxic spillover'.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
- Centre National de la Recherche Scientifique, 13000 Marseille, France
| | - Jean-Christophe Lagier
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
| |
Collapse
|
31
|
Kim HY, Kang HG, Kim HM, Jeong HJ. Expression of SARS-CoV-2 receptor angiotensin-converting enzyme 2 by activating protein-1 in human mast cells. Cell Immunol 2023; 386:104705. [PMID: 36898276 PMCID: PMC9985914 DOI: 10.1016/j.cellimm.2023.104705] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection activates mast cells and induces a cytokine storm, leading to severe Coronavirus disease in 2019 (COVID-19). SARS-CoV-2 employs angiotensin-converting enzyme 2 (ACE2) for cell entry. In the present study, the expression of ACE2 and its mechanism in activated mast cells were studied utilizing the human mast cell line, HMC-1 cells and it was elucidated whether dexamethasone used as a treatment for COVID-19 could regulate ACE2 expression. Here we documented for the first time that levels of ACE2 were increased by stimulation of phorbol 12-myristate 13-acetate and A23187 (PMACI) in HMC-1 cells. Increased levels of ACE2 were significantly diminished by treatment with Wortmannin, SP600125, SB203580, PD98059, or SR11302. The expression of ACE2 was most significantly reduced by the activating protein (AP)-1 inhibitor SR11302. PMACI stimulation enhanced the expression of the transcription factor AP-1 for ACE2. In addition, levels of transmembrane protease/serine subfamily member 2 (TMPRSS2) and tryptase were increased in PMACI-stimulated HMC-1 cells. However, dexamethasone significantly lowered levels of ACE2, TMPRSS2, and tryptase generated by PMACI. Treatment with dexamethasone also reduced activation of signaling molecules linked to ACE2 expression. According to these findings, levels of ACE2 were up-regulated through activation of AP-1 in mast cells, suggesting that suppressing ACE2 levels in mast cells would be a therapeutic approach to lessen the harm caused by COVID-19.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Biochip Research Center, Hoseo University, Asan 31499, Republic of Korea
| | - Ho-Geun Kang
- Department of Bio-Convergence System, Graduate School, Hoseo University, Asan 31499, Republic of Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea.
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan 31499, Republic of Korea; Department of Bio-Convergence System, Graduate School, Hoseo University, Asan 31499, Republic of Korea.
| |
Collapse
|
32
|
Porter LM, Guo W, Crozier TWM, Greenwood EJD, Ortmann B, Kottmann D, Nathan JA, Mahadeva R, Lehner PJ, McCaughan F. Cigarette smoke preferentially induces full length ACE2 expression in differentiated primary human airway cultures but does not alter the efficiency of cellular SARS-CoV-2 infection. Heliyon 2023; 9:e14383. [PMID: 36938474 PMCID: PMC10005841 DOI: 10.1016/j.heliyon.2023.e14383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Cigarette smoking has many serious negative health consequences. The relationship between smoking and SARS-CoV-2 infection is controversial, specifically whether smokers are at increased risk of infection. We investigated the impact of cigarette smoke on ACE2 isoform expression and SARS-CoV-2 infection in differentiated primary human bronchial epithelial cells at the air-liquid-interface (ALI). We assessed the expression of ACE2 in response to CSE and therapeutics reported to modulate ACE2. We exposed ALI cultures to cigarette smoke extract (CSE) and then infected them with SARS-CoV-2. We measured cellular infection using flow cytometry and whole-transwell immunofluorescence. We found that CSE increased expression of full-length ACE2 (flACE2) but did not alter the expression of a Type I-interferon sensitive truncated isoform (dACE2) that lacks the capacity to bind SARS-CoV-2. CSE did not have a significant impact on key mediators of the innate immune response. Importantly, we show that, despite the increase in flACE2, CSE did not alter airway cell infection after CSE exposure. We found that nicotine does not significantly alter flACE2 expression but that NRF2 agonists do lead to an increase in flACE2 expression. This increase was not associated with an increase in SARS-CoV-2 infection. Our results are consistent with the epidemiological data suggesting that current smokers do not have an excess of SARS-CoV-2 infection. but that those with chronic respiratory or cardiovascular disease are more vulnerable to severe COVID-19. They suggest that, in differentiated conducting airway cells, flACE2 expression levels may not limit airway SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Linsey M. Porter
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, CB2 OQQ, UK
| | - Wenrui Guo
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, CB2 OQQ, UK
| | - Thomas WM. Crozier
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Edward JD. Greenwood
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Brian Ortmann
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Daniel Kottmann
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, CB2 OQQ, UK
| | - James A. Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Ravindra Mahadeva
- Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Addenbrookes Hospital, Cambridge, CB2 OQQ, UK
| | - Paul J. Lehner
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Frank McCaughan
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, CB2 OQQ, UK
| |
Collapse
|
33
|
Beijnen EMS, Odumade OA, Haren SDV. Molecular Determinants of the Early Life Immune Response to COVID-19 Infection and Immunization. Vaccines (Basel) 2023; 11:vaccines11030509. [PMID: 36992093 DOI: 10.3390/vaccines11030509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/11/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Clinical manifestations from primary COVID infection in children are generally less severe as compared to adults, and severe pediatric cases occur predominantly in children with underlying medical conditions. However, despite the lower incidence of disease severity, the burden of COVID-19 in children is not negligible. Throughout the course of the pandemic, the case incidence in children has substantially increased, with estimated cumulative rates of SARS-CoV-2 infection and COVID-19 symptomatic illness in children comparable to those in adults. Vaccination is a key approach to enhance immunogenicity and protection against SARS-CoV-2. Although the immune system of children is functionally distinct from that of other age groups, vaccine development specific for the pediatric population has mostly been limited to dose-titration of formulations that were developed primarily for adults. In this review, we summarize the literature pertaining to age-specific differences in COVID-19 pathogenesis and clinical manifestation. In addition, we review molecular distinctions in how the early life immune system responds to infection and vaccination. Finally, we discuss recent advances in development of pediatric COVID-19 vaccines and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M S Beijnen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Oludare A Odumade
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Division of Medicine Critical Care, Boston Children's Hospital, Boston, MA 02115, USA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Lapo-Talledo GJ, Talledo-Delgado JA, Fernández-Aballí LS. A competing risk survival analysis of the sociodemographic factors of COVID-19 in-hospital mortality in Ecuador. CAD SAUDE PUBLICA 2023; 39:e00294721. [PMID: 36753093 DOI: 10.1590/0102-311xen294721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/01/2022] [Indexed: 02/09/2023] Open
Abstract
This study aimed to analyze the effect of sociodemographic characteristics on COVID-19 in-hospital mortality in Ecuador from March 1 to December 31, 2020. This retrospective longitudinal study was performed with data from publicly accessible registries of the Ecuadorian National Institute of Statistics and Censuses (INEC). Data underwent a competing risk analysis with estimates of the cumulative incidence function (CIF). The effect of covariates on CIFs was estimated using the Fine-Gray model and results were expressed as adjusted subdistribution hazard ratios (SHR). The analysis included 30,991 confirmed COVID-19 patients with a mean age of 56.57±18.53 years; 60.7% (n = 18,816) were men and 39.3% (n = 12,175) were women. Being of advanced age, especially older than or equal to 75 years (SHR = 17.97; 95%CI: 13.08-24.69), being a man (SHR = 1.29; 95%CI: 1.22-1.36), living in rural areas (SHR = 1.18; 95%CI: 1.10-1.26), and receiving care in a public health center (SHR = 1.64; 95%CI: 1.51-1.78) were factors that increased the incidence of death from COVID-19, while living at an elevation higher than 2,500 meters above sea level (SHR = 0.69; 95%CI: 0.66-0.73) decreased this incidence. Since the incidence of death for individuals living in rural areas and who received medical care from the public sector was higher, income and poverty are important factors in the final outcome of this disease.
Collapse
|
35
|
Bellavite P, Ferraresi A, Isidoro C. Immune Response and Molecular Mechanisms of Cardiovascular Adverse Effects of Spike Proteins from SARS-CoV-2 and mRNA Vaccines. Biomedicines 2023; 11:451. [PMID: 36830987 PMCID: PMC9953067 DOI: 10.3390/biomedicines11020451] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
The SARS-CoV-2 (severe acute respiratory syndrome coronavirus responsible for the COVID-19 disease) uses the Spike proteins of its envelope for infecting target cells expressing on the membrane the angiotensin converting enzyme 2 (ACE2) enzyme that acts as a receptor. To control the pandemic, genetically engineered vaccines have been designed for inducing neutralizing antibodies against the Spike proteins. These vaccines do not act like traditional protein-based vaccines, as they deliver the message in the form of mRNA or DNA to host cells that then produce and expose the Spike protein on the membrane (from which it can be shed in soluble form) to alert the immune system. Mass vaccination has brought to light various adverse effects associated with these genetically based vaccines, mainly affecting the circulatory and cardiovascular system. ACE2 is present as membrane-bound on several cell types, including the mucosa of the upper respiratory and of the gastrointestinal tracts, the endothelium, the platelets, and in soluble form in the plasma. The ACE2 enzyme converts the vasoconstrictor angiotensin II into peptides with vasodilator properties. Here we review the pathways for immunization and the molecular mechanisms through which the Spike protein, either from SARS-CoV-2 or encoded by the mRNA-based vaccines, interferes with the Renin-Angiotensin-System governed by ACE2, thus altering the homeostasis of the circulation and of the cardiovascular system. Understanding the molecular interactions of the Spike protein with ACE2 and the consequent impact on cardiovascular system homeostasis will direct the diagnosis and therapy of the vaccine-related adverse effects and provide information for development of a personalized vaccination that considers pathophysiological conditions predisposing to such adverse events.
Collapse
Affiliation(s)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
36
|
Tang AT, Buchholz DW, Szigety KM, Imbiakha B, Gao S, Frankfurter M, Wang M, Yang J, Hewins P, Mericko-Ishizuka P, Leu NA, Sterling S, Monreal IA, Sahler J, August A, Zhu X, Jurado KA, Xu M, Morrisey EE, Millar SE, Aguilar HC, Kahn ML. Cell-autonomous requirement for ACE2 across organs in lethal mouse SARS-CoV-2 infection. PLoS Biol 2023; 21:e3001989. [PMID: 36745682 PMCID: PMC9934376 DOI: 10.1371/journal.pbio.3001989] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/16/2023] [Accepted: 01/04/2023] [Indexed: 02/07/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the cell-surface receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). While its central role in Coronavirus Disease 2019 (COVID-19) pathogenesis is indisputable, there remains significant debate regarding the role of this transmembrane carboxypeptidase in the disease course. These include the role of soluble versus membrane-bound ACE2, as well as ACE2-independent mechanisms that may contribute to viral spread. Testing these roles requires in vivo models. Here, we report humanized ACE2-floxed mice in which hACE2 is expressed from the mouse Ace2 locus in a manner that confers lethal disease and permits cell-specific, Cre-mediated loss of function, and LSL-hACE2 mice in which hACE2 is expressed from the Rosa26 locus enabling cell-specific, Cre-mediated gain of function. Following exposure to SARS-CoV-2, hACE2-floxed mice experienced lethal cachexia, pulmonary infiltrates, intravascular thrombosis and hypoxemia-hallmarks of severe COVID-19. Cre-mediated loss and gain of hACE2 demonstrate that neuronal infection confers lethal cachexia, hypoxemia, and respiratory failure in the absence of lung epithelial infection. In this series of genetic experiments, we demonstrate that ACE2 is absolutely and cell-autonomously required for SARS-CoV-2 infection in the olfactory epithelium, brain, and lung across diverse cell types. Therapies inhibiting or blocking ACE2 at these different sites are likely to be an effective strategy towards preventing severe COVID-19.
Collapse
Affiliation(s)
- Alan T. Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David W. Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Katherine M. Szigety
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brian Imbiakha
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Siqi Gao
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maxwell Frankfurter
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Min Wang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Peter Hewins
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Patricia Mericko-Ishizuka
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - N Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Isaac A. Monreal
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Julie Sahler
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Xuming Zhu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kellie A. Jurado
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mingang Xu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Edward E. Morrisey
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Sarah E. Millar
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Mark L. Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
37
|
Russell CD, Lone NI, Baillie JK. Comorbidities, multimorbidity and COVID-19. Nat Med 2023; 29:334-343. [PMID: 36797482 DOI: 10.1038/s41591-022-02156-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/25/2022] [Indexed: 02/18/2023]
Abstract
The influence of comorbidities on COVID-19 outcomes has been recognized since the earliest days of the pandemic. But establishing causality and determining underlying mechanisms and clinical implications has been challenging-owing to the multitude of confounding factors and patient variability. Several distinct pathological mechanisms, not active in every patient, determine health outcomes in the three different phases of COVID-19-from the initial viral replication phase to inflammatory lung injury and post-acute sequelae. Specific comorbidities (and overall multimorbidity) can either exacerbate these pathological mechanisms or reduce the patient's tolerance to organ injury. In this Review, we consider the impact of specific comorbidities, and overall multimorbidity, on the three mechanistically distinct phases of COVID-19, and we discuss the utility of host genetics as a route to causal inference by eliminating many sources of confounding. Continued research into the mechanisms of disease-state interactions will be crucial to inform stratification of therapeutic approaches and improve outcomes for patients.
Collapse
Affiliation(s)
- Clark D Russell
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Nazir I Lone
- Usher Institute, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK.
- Intensive Care Unit, Royal Infirmary of Edinburgh, Little France Crescent, Edinburgh, UK.
| | - J Kenneth Baillie
- Intensive Care Unit, Royal Infirmary of Edinburgh, Little France Crescent, Edinburgh, UK.
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK.
- Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, UK.
| |
Collapse
|
38
|
Valyaeva AA, Zharikova AA, Sheval EV. SARS-CoV-2 cellular tropism and direct multiorgan failure in COVID-19 patients: Bioinformatic predictions, experimental observations, and open questions. Cell Biol Int 2023; 47:308-326. [PMID: 36229927 PMCID: PMC9874490 DOI: 10.1002/cbin.11928] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/24/2022] [Accepted: 09/25/2022] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), has led to an unprecedented public health emergency worldwide. While common cold symptoms are observed in mild cases, COVID-19 is accompanied by multiorgan failure in severe patients. Organ damage in COVID-19 patients is partially associated with the indirect effects of SARS-CoV-2 infection (e.g., systemic inflammation, hypoxic-ischemic damage, coagulopathy), but early processes in COVID-19 patients that trigger a chain of indirect effects are connected with the direct infection of cells by the virus. To understand the virus transmission routes and the reasons for the wide-spectrum of complications and severe outcomes of COVID-19, it is important to identify the cells targeted by SARS-CoV-2. This review summarizes the major steps of investigation and the most recent findings regarding SARS-CoV-2 cellular tropism and the possible connection between the early stages of infection and multiorgan failure in COVID-19. The SARS-CoV-2 pandemic is the first epidemic in which data extracted from single-cell RNA-seq (scRNA-seq) gene expression data sets have been widely used to predict cellular tropism. The analysis presented here indicates that the SARS-CoV-2 cellular tropism predictions are accurate enough for estimating the potential susceptibility of different cells to SARS-CoV-2 infection; however, it appears that not all susceptible cells may be infected in patients with COVID-19.
Collapse
Affiliation(s)
- Anna A. Valyaeva
- School of Bioengineering and BioinformaticsLomonosov Moscow State UniversityMoscowRussia
- Belozersky Institute of Physico‐Chemical BiologyLomonosov Moscow State UniversityMoscowRussia
| | - Anastasia A. Zharikova
- School of Bioengineering and BioinformaticsLomonosov Moscow State UniversityMoscowRussia
- Belozersky Institute of Physico‐Chemical BiologyLomonosov Moscow State UniversityMoscowRussia
| | - Eugene V. Sheval
- School of Bioengineering and BioinformaticsLomonosov Moscow State UniversityMoscowRussia
- Belozersky Institute of Physico‐Chemical BiologyLomonosov Moscow State UniversityMoscowRussia
- Department of Cell Biology and Histology, School of BiologyLomonosov Moscow State UniversityMoscowRussia
| |
Collapse
|
39
|
Meyerholz DK. Rigid respiration: fulminant pulmonary fibrosis after COVID-19. EBioMedicine 2023; 87:104428. [PMID: 36580850 PMCID: PMC9792276 DOI: 10.1016/j.ebiom.2022.104428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Affiliation(s)
- David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
40
|
Hönzke K, Obermayer B, Mache C, Fatykhova D, Kessler M, Dökel S, Wyler E, Baumgardt M, Löwa A, Hoffmann K, Graff P, Schulze J, Mieth M, Hellwig K, Demir Z, Biere B, Brunotte L, Mecate-Zambrano A, Bushe J, Dohmen M, Hinze C, Elezkurtaj S, Tönnies M, Bauer TT, Eggeling S, Tran HL, Schneider P, Neudecker J, Rückert JC, Schmidt-Ott KM, Busch J, Klauschen F, Horst D, Radbruch H, Radke J, Heppner F, Corman VM, Niemeyer D, Müller MA, Goffinet C, Mothes R, Pascual-Reguant A, Hauser AE, Beule D, Landthaler M, Ludwig S, Suttorp N, Witzenrath M, Gruber AD, Drosten C, Sander LE, Wolff T, Hippenstiel S, Hocke AC. Human lungs show limited permissiveness for SARS-CoV-2 due to scarce ACE2 levels but virus-induced expansion of inflammatory macrophages. Eur Respir J 2022; 60:2102725. [PMID: 35728978 PMCID: PMC9712848 DOI: 10.1183/13993003.02725-2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilises the angiotensin-converting enzyme 2 (ACE2) transmembrane peptidase as cellular entry receptor. However, whether SARS-CoV-2 in the alveolar compartment is strictly ACE2-dependent and to what extent virus-induced tissue damage and/or direct immune activation determines early pathogenesis is still elusive. METHODS Spectral microscopy, single-cell/-nucleus RNA sequencing or ACE2 "gain-of-function" experiments were applied to infected human lung explants and adult stem cell derived human lung organoids to correlate ACE2 and related host factors with SARS-CoV-2 tropism, propagation, virulence and immune activation compared to SARS-CoV, influenza and Middle East respiratory syndrome coronavirus (MERS-CoV). Coronavirus disease 2019 (COVID-19) autopsy material was used to validate ex vivo results. RESULTS We provide evidence that alveolar ACE2 expression must be considered scarce, thereby limiting SARS-CoV-2 propagation and virus-induced tissue damage in the human alveolus. Instead, ex vivo infected human lungs and COVID-19 autopsy samples showed that alveolar macrophages were frequently positive for SARS-CoV-2. Single-cell/-nucleus transcriptomics further revealed nonproductive virus uptake and a related inflammatory and anti-viral activation, especially in "inflammatory alveolar macrophages", comparable to those induced by SARS-CoV and MERS-CoV, but different from NL63 or influenza virus infection. CONCLUSIONS Collectively, our findings indicate that severe lung injury in COVID-19 probably results from a macrophage-triggered immune activation rather than direct viral damage of the alveolar compartment.
Collapse
Affiliation(s)
- Katja Hönzke
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Contributed equally
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
- Contributed equally
| | - Christin Mache
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
- Contributed equally
| | - Diana Fatykhova
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mirjana Kessler
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gynecology and Obstetrics, Ludwig-Maximilian University, Munich, Germany
| | - Simon Dökel
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Morris Baumgardt
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Löwa
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karen Hoffmann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Patrick Graff
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jessica Schulze
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Maren Mieth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Hellwig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Zeynep Demir
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Biere
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Linda Brunotte
- Institute of Virology, Westfaelische Wilhelms Universität, Münster, Germany
| | | | - Judith Bushe
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Melanie Dohmen
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mario Tönnies
- HELIOS Clinic Emil von Behring, Department of Pneumology and Department of Thoracic Surgery, Chest Hospital Heckeshorn, Berlin, Germany
| | - Torsten T Bauer
- HELIOS Clinic Emil von Behring, Department of Pneumology and Department of Thoracic Surgery, Chest Hospital Heckeshorn, Berlin, Germany
| | - Stephan Eggeling
- Department of Thoracic Surgery, Vivantes Clinics Neukölln, Berlin, Germany
| | - Hong-Linh Tran
- Department of Thoracic Surgery, Vivantes Clinics Neukölln, Berlin, Germany
| | - Paul Schneider
- Department for Thoracic Surgery, DRK Clinics, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jens C Rückert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jonas Busch
- Clinic for Urology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederick Klauschen
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Helena Radbruch
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Josefine Radke
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank Heppner
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniela Niemeyer
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ronja Mothes
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Anna Pascual-Reguant
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Stephan Ludwig
- Institute of Virology, Westfaelische Wilhelms Universität, Münster, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif-Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thorsten Wolff
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas C Hocke
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
41
|
Abrehart T, Suryadinata R, McCafferty C, Jacobson J, Ignjatovic V, Robinson P, Crawford NW, Monagle P, Subbarao K, Satzke C, Wurzel D. Age-related differences in SARS-CoV-2 binding factors: An explanation for reduced susceptibility to severe COVID-19 among children? Paediatr Respir Rev 2022; 44:61-69. [PMID: 35227628 PMCID: PMC8823960 DOI: 10.1016/j.prrv.2022.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023]
Abstract
CONTEXT In contrast with other respiratory viruses, children infected with SARS-CoV-2 are largely spared from severe COVID-19. OBJECTIVES To critically assess age-related differences in three host proteins involved in SARS-CoV-2 cellular entry: angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) and furin. METHODS We systematically searched Medline, Embase, and PubMed databases for relevant publications. Studies were eligible if they evaluated ACE2, TMPRSS2 or furin expression, methylation, or protein level in children. RESULTS Sixteen papers were included. Age-dependent differences in membrane-bound and soluble ACE2 were shown in several studies, with ACE2 expression increasing with age. TMPRSS2 and furin are key proteases involved in SARS-CoV-2 spike protein cleavage. TMPRSS2 expression is increased by circulating androgens and is thus low in pre-pubertal children. Furin has not currently been well researched. LIMITATIONS High levels of study heterogeneity. CONCLUSIONS Low expression of key host proteins may partially explain the reduced incidence of severe COVID-19 among children, although further research is needed.
Collapse
Affiliation(s)
- Thomas Abrehart
- Department of Paediatrics, the University of Melbourne, Parkville, Australia.
| | - Randy Suryadinata
- Department of Respiratory Medicine, Royal Children’s Hospital, Melbourne, Victoria, Australia,Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia
| | - Conor McCafferty
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Haematology, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Jonathan Jacobson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia,Department of Microbiology and Immunology, the University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Vera Ignjatovic
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Haematology, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Phil Robinson
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Department of Respiratory Medicine, Royal Children’s Hospital, Melbourne, Victoria, Australia,Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia
| | - Nigel W. Crawford
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia
| | - Paul Monagle
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Haematology, Murdoch Children’s Research Institute, Melbourne, Australia,Department of Haematology, Royal Children’s Hospital, Parkville, Australia,Kids Cancer Centre, Sydney Children’s Hospital, Randwick, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia,Department of Microbiology and Immunology, the University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Catherine Satzke
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia,Department of Microbiology and Immunology, the University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Danielle Wurzel
- Department of Paediatrics, the University of Melbourne, Parkville, Australia,Department of Respiratory Medicine, Royal Children’s Hospital, Melbourne, Victoria, Australia,Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia,Melbourne School of Population and Global Health, the University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. Int J Mol Sci 2022; 23:ijms232315122. [PMID: 36499448 PMCID: PMC9737069 DOI: 10.3390/ijms232315122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
COVID-19, a significant global health threat, appears to be an immune-related disease. Failure of effective immune responses in initial stages of infection may contribute to development of cytokine storm and systemic inflammation with organ damage, leading to poor clinical outcomes. Disease severity and the emergence of new SARS-CoV-2 variants highlight the need for new preventative and therapeutic strategies to protect the immunocompromised population. Available data indicate that these people may benefit from adoptive transfer of allogeneic SARS-CoV-2-specific T cells isolated from convalescent individuals. This review first provides an insight into the mechanism of cytokine storm development, as it is directly related to the exhaustion of T cell population, essential for viral clearance and long-term antiviral immunity. Next, we describe virus-specific T lymphocytes as a promising and efficient approach for the treatment and prevention of severe COVID-19. Furthermore, other potential cell-based therapies, including natural killer cells, regulatory T cells and mesenchymal stem cells are mentioned. Additionally, we discuss fast and effective ways of producing clinical-grade antigen-specific T cells which can be cryopreserved and serve as an effective "off-the-shelf" approach for rapid treatment of SARS-CoV-2 infection in case of sudden patient deterioration.
Collapse
|
43
|
Devaux CA, Camoin-Jau L. An update on angiotensin-converting enzyme 2 structure/functions, polymorphism, and duplicitous nature in the pathophysiology of coronavirus disease 2019: Implications for vascular and coagulation disease associated with severe acute respiratory syndrome coronavirus infection. Front Microbiol 2022; 13:1042200. [PMID: 36519165 PMCID: PMC9742611 DOI: 10.3389/fmicb.2022.1042200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 08/01/2023] Open
Abstract
It has been known for many years that the angiotensin-converting enzyme 2 (ACE2) is a cell surface enzyme involved in the regulation of blood pressure. More recently, it was proven that the severe acute respiratory syndrome coronavirus (SARS-CoV-2) interacts with ACE2 to enter susceptible human cells. This functional duality of ACE2 tends to explain why this molecule plays such an important role in the clinical manifestations of coronavirus disease 2019 (COVID-19). At the very start of the pandemic, a publication from our Institute (entitled "ACE2 receptor polymorphism: susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome"), was one of the first reviews linking COVID-19 to the duplicitous nature of ACE2. However, even given that COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin system (RAS), we were still far from understanding the complexity of the mechanisms which are controlled by ACE2 in different cell types. To gain insight into the physiopathology of SARS-CoV-2 infection, it is essential to consider the polymorphism and expression levels of the ACE2 gene (including its alternative isoforms). Over the past 2 years, an impressive amount of new results have come to shed light on the role of ACE2 in the pathophysiology of COVID-19, requiring us to update our analysis. Genetic linkage studies have been reported that highlight a relationship between ACE2 genetic variants and the risk of developing hypertension. Currently, many research efforts are being undertaken to understand the links between ACE2 polymorphism and the severity of COVID-19. In this review, we update the state of knowledge on the polymorphism of ACE2 and its consequences on the susceptibility of individuals to SARS-CoV-2. We also discuss the link between the increase of angiotensin II levels among SARS-CoV-2-infected patients and the development of a cytokine storm associated microvascular injury and obstructive thrombo-inflammatory syndrome, which represent the primary causes of severe forms of COVID-19 and lethality. Finally, we summarize the therapeutic strategies aimed at preventing the severe forms of COVID-19 that target ACE2. Changing paradigms may help improve patients' therapy.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Center National de la Recherche Scientifique, Marseille, France
| | - Laurence Camoin-Jau
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Laboratoire d’Hématologie, Hôpital de La Timone, APHM, Boulevard Jean-Moulin, Marseille, France
| |
Collapse
|
44
|
Wedel J, Steinmann P, Štrakl M, Hriberšek M, Cui Y, Ravnik J. Anatomy matters: The role of the subject-specific respiratory tract on aerosol deposition - A CFD study. COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 2022; 401:115372. [PMID: 35919629 PMCID: PMC9333481 DOI: 10.1016/j.cma.2022.115372] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The COVID-19 pandemic is one of the greatest challenges to humanity nowadays. COVID-19 virus can replicate in the host's larynx region, which is in contrast to other viruses that replicate in lungs only, i.e. SARS. This is conjectured to support a fast spread of COVID-19. However, there is sparse research in this field about quantitative comparison of virus load in the larynx for varying susceptible individuals. In this regard the lung geometry itself could influence the risk of reproducing more pathogens and consequently exhaling more virus. Disadvantageously, there are only sparse lung geometries available. To still be able to investigate realistic geometrical deviations we employ three different digital replicas of human airways up to the 7 th level of bifurcation, representing two realistic lungs (male and female) as well as a more simplified experimental model. Our aim is to investigate the influence of breathing scenarios on aerosol deposition in anatomically different, realistic human airways. In this context, we employ three levels of cardiovascular activity as well as reported experimental particle size distributions by means of Computational Fluid Dynamics (CFD) with special focus on the larynx region to enable new insights into the local virus loads in human respiratory tracts. In addition, the influence of more realistic boundary conditions is investigated by performing transient simulations of a complete respiratory cycle in the upper lung regions of the considered respiratory models, focusing in particular on deposition in the oral cavity, the laryngeal region, and trachea, while simplifying the tracheobronchial tree. The aerosol deposition is modeled via OpenFOAM\protect \relax \special {t4ht=®} by employing an Euler-Lagrangian frame including steady and unsteady Reynolds Averaged Navier-Stokes (RANS) resolved turbulent flow using the k- ω -SST and k- ω -SST DES turbulence models. We observed that the respiratory geometry altered the local deposition patterns, especially in the laryngeal region. Despite the larynx region, the effects of varying flow rate for the airway geometries considered were found to be similar in the majority of respiratory tract regions. For all particle size distributions considered, localized particle accumulation occurred in the larynx of all considered lung models, which were more pronounced for larger particle size distributions. Moreover, it was found, that employing transient simulations instead of steady-state analysis, the overall particle deposition pattern is maintained, however with a stronger intensity in the transient cases.
Collapse
Affiliation(s)
- Jana Wedel
- Institute of Applied Mechanics, Universität Erlangen-Nürnberg, Germany
| | - Paul Steinmann
- Institute of Applied Mechanics, Universität Erlangen-Nürnberg, Germany
- Glasgow Computational Engineering Center, University of Glasgow, United Kingdom
| | - Mitja Štrakl
- Faculty of Mechanical Engineering, University of Maribor, Slovenia
| | - Matjaž Hriberšek
- Faculty of Mechanical Engineering, University of Maribor, Slovenia
| | - Yan Cui
- Huazhong University of Science and Technology, Wuhan, China
| | - Jure Ravnik
- Faculty of Mechanical Engineering, University of Maribor, Slovenia
| |
Collapse
|
45
|
Hydroxychloroquine blocks SARS-CoV-2 entry into the endocytic pathway in mammalian cell culture. Commun Biol 2022; 5:958. [PMID: 36104427 PMCID: PMC9472185 DOI: 10.1038/s42003-022-03841-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Hydroxychloroquine (HCQ), a drug used to treat lupus and malaria, was proposed as a treatment for SARS-coronavirus-2 (SARS-CoV-2) infection, albeit with controversy. In vitro, HCQ effectively inhibits viral entry, but its use in the clinic has been hampered by conflicting results. A better understanding of HCQ’s mechanism of actions in vitro is needed. Recently, anesthetics were shown to disrupt ordered clusters of monosialotetrahexosylganglioside1 (GM1) lipid. These same lipid clusters recruit the SARS-CoV-2 surface receptor angiotensin converting enzyme 2 (ACE2) to endocytic lipids, away from phosphatidylinositol 4,5 bisphosphate (PIP2) clusters. Here we employed super-resolution imaging of cultured mammalian cells (VeroE6, A549, H1793, and HEK293T) to show HCQ directly perturbs clustering of ACE2 receptor with both endocytic lipids and PIP2 clusters. In elevated (high) cholesterol, HCQ moves ACE2 nanoscopic distances away from endocytic lipids. In cells with resting (low) cholesterol, ACE2 primarily associates with PIP2 clusters, and HCQ moves ACE2 away from PIP2 clusters—erythromycin has a similar effect. We conclude HCQ inhibits viral entry through two distinct mechanisms in high and low tissue cholesterol and does so prior to inhibiting cathepsin-L. HCQ clinical trials and animal studies will need to account for tissue cholesterol levels when evaluating dosing and efficacy. Super-resolution microscopy in cultured cells is employed to dissect the effect of hydroxychloroquine (HCQ) at the plasma membrane and HCQ directly perturbs clustering of the SARS-CoV-2 receptor ACE2 with endocytic lipids and PIP2 clusters.
Collapse
|
46
|
Tran HTT, Peterburs P, Seibel J, Abramov-Sommariva D, Lamy E. In vitro Screening of Herbal Medicinal Products for Their Supportive Curing Potential in the Context of SARS-CoV-2. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8038195. [PMID: 36110194 PMCID: PMC9470301 DOI: 10.1155/2022/8038195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/06/2022] [Accepted: 07/23/2022] [Indexed: 11/17/2022]
Abstract
COVID-19 herbal medicinal products may have the potential for symptom relief in nonsevere or moderate disease cases. In this in vitro study we screened the five herbal medicinal products Sinupret extract (SINx), Bronchipret thyme-ivy (BRO-TE), Bronchipret thyme-primula (BRO TP), Imupret (IMU), and Tonsipret (TOP) with regard to their potential to (i) interfere with the binding of the human angiotensin-converting enzyme 2 (ACE2) receptor with the SARS-CoV-2 spike S1 protein, (ii) modulate the release of the human defensin HBD1 and cathelicidin LL-37 from human A549 lung cells upon spike S1 protein stimulation, and (iii) modulate the release of IFN-γ from activated human peripheral blood mononuclear cells (PBMC). The effect of the extracts on the interaction of spike S1 protein and the human ACE2 receptor was measured by ELISA. The effects on the intracellular IFN-γ expression in stimulated human PBMC were measured by flow cytometry. Regulation of HBD1 and LL-37 expression and secretion was assessed in 25 d long-term cultured human lung A549 epithelial cells by RT-PCR and ELISA. IMU and BRO-TE concentration-dependently inhibited the interaction between spike S1 protein and the ACE2 receptor. SINx, TOP, and BRO-TE significantly upregulated the intracellular expression of anti-viral IFN-γ from stimulated PBMC. Cotreatment of A549 cells with IMU or BRO TP together with SARS-CoV-2 spike protein significantly upregulated mRNA expression (IMU) and release of HBD1 (IMU and BRO TP) and LL-37 (BRO TP). The in vitro screening results provide first evidence for an immune-activating potential of some of the tested herbal medicinal extracts in the context of SARS-CoV-2. Whether these could be supportive in symptom relief or curing from SARS-CoV-2 infection needs deeper understanding of the observations.
Collapse
Affiliation(s)
- Hoai Thi Thu Tran
- Molecular Preventive Medicine, University of Freiburg, Medical Center and Faculty of Medicine, Engesserstraße 4, 79108 Freiburg, Germany
| | | | - Jan Seibel
- Bionorica SE, Kerschensteinerstraße 11-15, 92318 Neumarkt, Germany
| | | | - Evelyn Lamy
- Molecular Preventive Medicine, University of Freiburg, Medical Center and Faculty of Medicine, Engesserstraße 4, 79108 Freiburg, Germany
| |
Collapse
|
47
|
Izzo R, Trimarco V, Mone P, Aloè T, Capra Marzani M, Diana A, Fazio G, Mallardo M, Maniscalco M, Marazzi G, Messina N, Mininni S, Mussi C, Pelaia G, Pennisi A, Santus P, Scarpelli F, Tursi F, Zanforlin A, Santulli G, Trimarco B. Combining L-Arginine with vitamin C improves long-COVID symptoms: The LINCOLN Survey. Pharmacol Res 2022; 183:106360. [PMID: 35868478 PMCID: PMC9295384 DOI: 10.1016/j.phrs.2022.106360] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Recent evidence suggests that oxidative stress and endothelial dysfunction play critical roles in the pathophysiology of COVID-19 and Long-COVID. We hypothesized that a supplementation combining L-Arginine (to improve endothelial function) and Vitamin C (to reduce oxidation) could have favorable effects on Long-COVID symptoms. METHODS We designed a survey (LINCOLN: L-Arginine and Vitamin C improves Long-COVID), assessing several symptoms that have been associated with Long-COVID to be administered nationwide to COVID-19 survivors; the survey also included effort perception, measured using the Borg scale. Patients receiving the survey were divided in two groups, with a 2:1 ratio: the first group included patients that received L-Arginine + Vitamin C, whereas the second group received a multivitamin combination (alternative treatment). RESULTS 1390 patients successfully completed the survey. Following a 30-day treatment in both groups, the survey revealed that patients in the L-Arginine + Vitamin C treatment arm had significantly lower scores compared to patients who had received the multivitamin combination. There were no other significant differences between the two groups. When examining effort perception, we observed a significantly lower value (p < 0.0001) in patients receiving L-Arginine + Vitamin C compared to the alternative-treatment arm. CONCLUSIONS Our survey indicates that the supplementation with L-Arginine + Vitamin C has beneficial effects in Long-COVID, in terms of attenuating its typical symptoms and improving effort perception.
Collapse
Affiliation(s)
- Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University, Naples, Italy
| | - Pasquale Mone
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | - Simone Mininni
- Associazione Scientifica Interdisciplinare Aggiornamento Medico (ASIAM), Florence, Italy
| | - Chiara Mussi
- Department of Biomedical and Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy
| | - Girolamo Pelaia
- Department of Health Science, Magna Graecia University, Catanzaro, Italy
| | | | | | | | | | | | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
48
|
McSweeney MD, Stewart I, Richardson Z, Kang H, Park Y, Kim C, Tiruthani K, Wolf W, Schaefer A, Kumar P, Aurora H, Hutchins J, Cho JM, Hickey AJ, Lee SY, Lai SK. Stable nebulization and muco-trapping properties of regdanvimab/IN-006 support its development as a potent, dose-saving inhaled therapy for COVID-19. Bioeng Transl Med 2022; 8:e10391. [PMID: 36248234 PMCID: PMC9537933 DOI: 10.1002/btm2.10391] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 01/21/2023] Open
Abstract
The respiratory tract represents the key target for antiviral delivery in early interventions to prevent severe COVID-19. While neutralizing monoclonal antibodies (mAb) possess considerable efficacy, their current reliance on parenteral dosing necessitates very large doses and places a substantial burden on the healthcare system. In contrast, direct inhaled delivery of mAb therapeutics offers the convenience of self-dosing at home, as well as much more efficient mAb delivery to the respiratory tract. Here, building on our previous discovery of Fc-mucin interactions crosslinking viruses to mucins, we showed that regdanvimab, a potent neutralizing mAb already approved for COVID-19 in several countries, can effectively trap SARS-CoV-2 virus-like particles in fresh human airway mucus. IN-006, a reformulation of regdanvimab, was stably nebulized across a wide range of concentrations, with no loss of activity and no formation of aggregates. Finally, nebulized delivery of IN-006 resulted in 100-fold greater mAb levels in the lungs of rats compared to serum, in marked contrast to intravenously dosed mAbs. These results not only support our current efforts to evaluate the safety and efficacy of IN-006 in clinical trials, but more broadly substantiate nebulized delivery of human antiviral mAbs as a new paradigm in treating SARS-CoV-2 and other respiratory pathologies.
Collapse
Affiliation(s)
- Morgan D. McSweeney
- Inhalon Biopharma IncResearch Triangle ParkNorth CarolinaUSA
- Mucommune LLCResearch Triangle ParkNorth CarolinaUSA
| | - Ian Stewart
- RTI InternationalResearch Triangle ParkNorth CarolinaUSA
| | - Zach Richardson
- Inhalon Biopharma IncResearch Triangle ParkNorth CarolinaUSA
- Mucommune LLCResearch Triangle ParkNorth CarolinaUSA
| | - Hyunah Kang
- Biotechnology Research InstituteCelltrion IncIncheonRepublic of Korea
| | - Yoona Park
- Biotechnology Research InstituteCelltrion IncIncheonRepublic of Korea
| | - Cheolmin Kim
- Biotechnology Research InstituteCelltrion IncIncheonRepublic of Korea
| | - Karthik Tiruthani
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
| | - Whitney Wolf
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
| | - Alison Schaefer
- UNC/NCSU Joint Department of Biomedical EngineeringUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
| | - Priya Kumar
- Department of Anesthesiology, School of MedicineUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Harendra Aurora
- Department of Anesthesiology, School of MedicineUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Jeff Hutchins
- Inhalon Biopharma IncResearch Triangle ParkNorth CarolinaUSA
| | - Jong Moon Cho
- Biotechnology Research InstituteCelltrion IncIncheonRepublic of Korea
| | | | - Soo Young Lee
- Biotechnology Research InstituteCelltrion IncIncheonRepublic of Korea
| | - Samuel K. Lai
- Inhalon Biopharma IncResearch Triangle ParkNorth CarolinaUSA
- Mucommune LLCResearch Triangle ParkNorth CarolinaUSA
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
- UNC/NCSU Joint Department of Biomedical EngineeringUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and Immunology, School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
49
|
Pandrea I, Brooks K, Desai RP, Tare M, Brenchley JM, Apetrei C. I've looked at gut from both sides now: Gastrointestinal tract involvement in the pathogenesis of SARS-CoV-2 and HIV/SIV infections. Front Immunol 2022; 13:899559. [PMID: 36032119 PMCID: PMC9411647 DOI: 10.3389/fimmu.2022.899559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/25/2022] [Indexed: 01/08/2023] Open
Abstract
The lumen of the gastrointestinal (GI) tract contains an incredibly diverse and extensive collection of microorganisms that can directly stimulate the immune system. There are significant data to demonstrate that the spatial localization of the microbiome can impact viral disease pathogenesis. Here we discuss recent studies that have investigated causes and consequences of GI tract pathologies in HIV, SIV, and SARS-CoV-2 infections with HIV and SIV initiating GI pathology from the basal side and SARS-CoV-2 from the luminal side. Both these infections result in alterations of the intestinal barrier, leading to microbial translocation, persistent inflammation, and T-cell immune activation. GI tract damage is one of the major contributors to multisystem inflammatory syndrome in SARS-CoV-2-infected individuals and to the incomplete immune restoration in HIV-infected subjects, even in those with robust viral control with antiretroviral therapy. While the causes of GI tract pathologies differ between these virus families, therapeutic interventions to reduce microbial translocation-induced inflammation and improve the integrity of the GI tract may improve the prognoses of infected individuals.
Collapse
Affiliation(s)
- Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rahul P. Desai
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Minali Tare
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
50
|
Faist A, Janowski J, Kumar S, Hinse S, Çalışkan DM, Lange J, Ludwig S, Brunotte L. Virus Infection and Systemic Inflammation: Lessons Learnt from COVID-19 and Beyond. Cells 2022; 11:2198. [PMID: 35883640 PMCID: PMC9316821 DOI: 10.3390/cells11142198] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/28/2022] [Accepted: 07/09/2022] [Indexed: 02/06/2023] Open
Abstract
Respiratory infections with newly emerging zoonotic viruses such as SARS-CoV-2, the etiological agent of COVID-19, often lead to the perturbation of the human innate and adaptive immune responses causing severe disease with high mortality. The responsible mechanisms are commonly virus-specific and often include either over-activated or delayed local interferon responses, which facilitate efficient viral replication in the primary target organ, systemic viral spread, and rapid onset of organ-specific and harmful inflammatory responses. Despite the distinct replication strategies, human infections with SARS-CoV-2 and highly pathogenic avian influenza viruses demonstrate remarkable similarities and differences regarding the mechanisms of immune induction, disease dynamics, as well as the long-term sequelae, which will be discussed in this review. In addition, we will highlight some important lessons about the effectiveness of antiviral and immunomodulatory therapeutic strategies that this pandemic has taught us.
Collapse
Affiliation(s)
- Aileen Faist
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- CiM-IMPRS, International Max Planck Research School—Molecular Biomedicine, Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany
| | - Josua Janowski
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- SP BioSciences Graduate Program, University of Muenster, 48149 Muenster, Germany
| | - Sriram Kumar
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
| | - Saskia Hinse
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
| | - Duygu Merve Çalışkan
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
| | - Julius Lange
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- CiM-IMPRS, International Max Planck Research School—Molecular Biomedicine, Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
- Interdisciplinary Center for Clinical Research, University of Muenster, 48149 Muenster, Germany
| | - Linda Brunotte
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- Interdisciplinary Center for Clinical Research, University of Muenster, 48149 Muenster, Germany
| |
Collapse
|